Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Drug Chem Toxicol ; 26(2): 99-115, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12816395

RESUMO

Methylglutaronitrile (MGN) is a high-boiling (263 degrees C) solvent/intermediate used in the fiber industry. Twenty male rats per group were exposed nose-only to condensation aerosol/vapor concentrations of approximately either 5, 25, or 200 mg/m3 of MGN for 6 h/day, 5 days/week over a 4-week period. Ten rats/group were sacrificed one day after the final exposure and the remaining rats after a four-week recovery period. No effects were observed in clinical observations during the exposure period, but body-weight depression was observed in the 200 mg/m3 group. The 200 mg/m3 group showed minimal decreases in red blood cell count, hemoglobin, and hematocrit values accompanied by increases in reticulocytes. There were no other effects observed in clinical or pathologic evaluations in the study. A neurobehavioral battery of tests (including grip strength, functional observational battery, and motor activity tests) given at the end of the exposure and recovery periods showed no MGN effects. During the 4-week recovery, body weights in the 200 mg/m3 group returned to normal and the hematologic findings in all groups were normal. Based on the above findings of body weight depression at 200 mg/m3, the no-observed-adverse-effect level (NOAEL) for this study was considered to be 25 mg/m3.


Assuntos
Glutaratos/toxicidade , Metemoglobina/análise , Nitrilas/toxicidade , Administração por Inalação , Animais , Câmaras de Exposição Atmosférica , Contagem de Células Sanguíneas , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Glutaratos/administração & dosagem , Masculino , Atividade Motora/efeitos dos fármacos , Sistema Nervoso/efeitos dos fármacos , Nitrilas/administração & dosagem , Nível de Efeito Adverso não Observado , Tamanho do Órgão/efeitos dos fármacos , Ratos , Fatores Sexuais , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica
2.
Drug Chem Toxicol ; 25(3): 255-66, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12173247

RESUMO

Some peroxisome proliferators have been reported to reduce body weight gain in suckling rats, possibly through a lactational effect. Decreases in milk production or nutritional quality, either as a result of peroxisome proliferator-induced reductions in lipid content or alterations in the hormonal milieu necessary for milk production, could result in pup growth retardation. Wyeth-14,643 (WY) is hypolipidemic agent and a potent inducer of hepatic peroxisome proliferation in rats and mice. As is commonly seen with rodent hepatic peroxisome proliferators, WY produces minimal or no peroxisome induction in guinea pigs or non-human primates. Goats are an excellent model for studying lactation, however, their sensitivity to peroxisome proliferating chemicals is not known. The present study was performed to assess the sensitivity of goats to the hypolipidemic and peroxisome proliferator properties of WY and to determine the effects of WY on milk quantity and quality. Six lactating adult female goats were assigned to either control or treated groups. Goats in the treated group were administered WY (40 mg/kg/day) for 14 consecutive days. The goats were milked twice daily in order to maintain lactation and the quantity of milk collected was recorded. Milk quality was evaluated by determining the content of total fat, protein, and carbohydrate in milk samples collected following 7 and 14 days of treatment. WY administration had no effects on final body weight, liver weight or, gross and histopathological findings. Milk quantity and quality were unaffected by treatment. Serum cholesterol and triglyceride levels were reduced by 25% compared to controls, although only the difference in cholesterol was statistically significant. Hepatic beta-oxidation (3 x control) and aromatase (1.5 x control) activities were significantly greater in the treatment group; however, there was no treatment-related effect in the total content of hepatic cytochrome P450. There was no difference in aromatase activity in a pooled ovarian microsome sample. Milk estradiol and prolactin concentrations were not affected by treatment. These findings indicate that goats are weak responders to the hepatic peroxisome proliferator effects of WY. Additionally, the slight serum hypolipidemic effect does not impact milk production or nutritional value.


Assuntos
Lactação/efeitos dos fármacos , Leite/química , Proliferadores de Peroxissomos/farmacologia , Pirimidinas/farmacologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Peso Corporal , Estradiol/análise , Feminino , Cabras , Lipídeos , Microcorpos , Ratos
3.
Toxicol Sci ; 67(2): 190-7, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12011478

RESUMO

Chronic administration of vinyl acetate (VA) in drinking water to rats and mice has produced upper digestive tract neoplasms. These tumors were believed to arise from the intracellular metabolism of VA by carboxylesterases to cytotoxic and genotoxic compounds. We hypothesized that prolonged VA exposure at high concentrations would induce cytotoxicity and a restorative cell proliferation (CP). These endpoints were measured in F-344 rats and BDF1 mice administered drinking water containing 0, 1000, 5000, 10,000, or 24,000 ppm VA for 92 days. On test days, Days 1, 8, 29, and 92, upper digestive tract histopathology and oral cavity CP (pulsed 5-bromodeoxyuridine [BrdU] to measure S-phase DNA synthesis) were evaluated. Analysis of test solutions showed that VA spontaneously hydrolyzed, slowly releasing acetic acid and thereby lowering pH. Statistically significant, concentration-related increases in CP occurred in basal cells of the mandibular oral cavity mucosa of mice at 10,000 and 24,000 ppm but only after 92 days. CP increases were approximately 2.4- and 3.4-fold above controls and were considered to be toxicologically significant. Some statistically significant increases in CP were also measured in the oral cavity mucosa of rats; however, these changes were considered to be of equivocal biological relevance. No histopathological evidence of mucosal injury was seen in either species. The absence of cytotoxicity in the upper digestive tract mucosa suggests that the increased CP at high administered VA concentrations may be due to a mitogenic response, ostensibly from the loss of cell growth controls in oral cavity mucosa.


Assuntos
Carcinógenos/toxicidade , Divisão Celular/efeitos dos fármacos , Compostos de Vinila/toxicidade , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Carcinógenos/administração & dosagem , DNA/biossíntese , Relação Dose-Resposta a Droga , Ingestão de Líquidos , Esôfago/efeitos dos fármacos , Esôfago/metabolismo , Esôfago/patologia , Mucosa Gástrica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/metabolismo , Mucosa Bucal/patologia , Ratos , Ratos Endogâmicos F344 , Estômago/efeitos dos fármacos , Estômago/patologia , Fatores de Tempo , Testes de Toxicidade , Compostos de Vinila/administração & dosagem , Abastecimento de Água
4.
Toxicol Sci ; 60(1): 44-55, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11222872

RESUMO

Wyeth-14,643 (WY) and ammonium perfluorooctanoate (C8) belong to a diverse class of compounds which have been shown to produce hepatic peroxisome proliferation in rodents. From previous work, WY, but not C8, has been shown to produce hepatocellular carcinoma in rats, while C8 has been shown to produce Leydig cell adenomas. In addition, based on a review of bioassay data a relationship appears to exist between peroxisome-proliferating compounds and Leydig cell adenoma and pancreatic acinar cell hyperplasia/adenocarcinoma formation. To further investigate the relationship between peroxisome-proliferating compounds and hepatic, Leydig cell, and pancreatic acinar cell tumorigenesis, a 2-year feeding study in male CD rats was initiated to test the hypothesis that peroxisome proliferating compounds induce a tumor triad (liver, Leydig cell, pancreatic acinar cell), and to examine the potential mechanism for the Leydig cell tumors. The study was conducted using 50 ppm WY and 300 ppm C8. The concentration of WY in the diet was decreased to 25 ppm on test day 301 due to increased mortality. In addition to the ad libitum control, a second control was pair-fed to the C8 group. Interim sacrifices were performed at 1- or 3-month intervals. Peroxisome proliferation measured by beta-oxidation activity and cell proliferation were measured in the liver and testis at all time points and in the pancreas beginning at the 9-month time point (cell proliferation only). Serum hormone concentrations (estradiol, testosterone, LH, FSH, and prolactin) were also measured at each time point. Increased relative liver weights and hepatic beta-oxidation activity were observed in both the WY- and C8-treated rats at all time points. In contrast, hepatic cell proliferation was significantly increased only in the WY-treated group. Neither WY nor C8 significantly altered the rate of Leydig cell beta-oxidation or Leydig cell proliferation when compared to the control groups. Moreover, the basal rate of beta-oxidation in Leydig cells was approximately 20 times less than the rate of hepatic beta-oxidation. There were no biologically meaningful differences in serum testosterone, FSH, prolactin, or LH concentrations in the WY- and C8-treated rats when compared to their respective controls. There were, however, significant increases in serum estradiol concentrations in the WY- and C8-treated rats at 1, 3, 6, 9, 15, 18, and 21 months. At 12 months, only the C8-treated rats had elevated serum estradiol concentrations when compared to the pair-fed control. Histopathological evaluation revealed compound-related increases in liver, Leydig cell, and pancreatic acinar cell tumors in both WY- and C8-treated rats. The data support the hypothesis that the peroxisome-proliferating compounds induce the previously described tumor triad. In addition, both C8 and WY produced a sustained increase in serum estradiol concentrations that correlated with the potency of the 2 compounds to induce Leydig cell tumors (i.e., WY caused a more consistent sustained increase in serum estradiol throughout the entire study, and more specifically at the end of the study, than did C8). This study suggests that estradiol may play a role in enhancement of Leydig cell tumors in the rat, and that peroxisome proliferators may induce tumors via a non-LH type mechanism.


Assuntos
Caprilatos/toxicidade , Carcinógenos/toxicidade , Fluorocarbonos/toxicidade , Neoplasias Experimentais/induzido quimicamente , Proliferadores de Peroxissomos/toxicidade , Pirimidinas/toxicidade , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/patologia , Animais , Testes de Carcinogenicidade , Divisão Celular/efeitos dos fármacos , Dieta , Estradiol/sangue , Hormônio Foliculoestimulante/sangue , Tumor de Células de Leydig/induzido quimicamente , Tumor de Células de Leydig/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/patologia , Longevidade/efeitos dos fármacos , Hormônio Luteinizante/sangue , Neoplasias Experimentais/sangue , Neoplasias Experimentais/patologia , Tamanho do Órgão/efeitos dos fármacos , Neoplasias Pancreáticas/induzido quimicamente , Neoplasias Pancreáticas/patologia , Prolactina/sangue , Ratos , Ratos Endogâmicos , Testosterona/sangue
5.
Reprod Toxicol ; 14(3): 193-205, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10838120

RESUMO

Apomorphine (APO; D(2) receptor agonist), haloperidol (HAL; D(2) receptor antagonist), and reserpine (RES; a dopamine depletor that acts to lower brain dopamine levels by depleting central nervous system monoamines via disrupting storage vesicle function) have been examined in a Tier I screening battery, which has been designed to detect endocrine-active compounds (EACs). The Tier I battery incorporates two short-term in vivo tests (a 5-day ovariectomized female battery and a 15-day intact male battery using Sprague-Dawley rats) and an in vitro yeast transactivation system (YTS). In addition, two blood collection procedures were evaluated for their utility in detecting HAL-induced increases in serum prolactin (PRL) levels (i.e., the stress associated with each procedure). In the in vivo female battery, both HAL and RES increased serum PRL concentrations as expected, although the increase caused by RES was marginal. Increases in serum PRL levels are enhanced when daily dosages are administered via multiple-daily dosing of the test compound, which results in higher sustained blood levels of the test compounds. APO failed to decrease serum PRL concentrations in the female battery. In the in vivo male battery, HAL increased serum PRL concentrations as expected. However, APO and RES failed to affect serum PRL concentrations. The blood collection comparison experiment demonstrated that possible confounding of the data can occur with serum PRL concentrations when animals are exposed to stress. Basal levels of PRL were approximately fourfold higher in animals that were bled via the tail vein procedure when compared to PRL levels from animals that were bled under CO(2) anesthesia at euthanization. As a result of the higher basal PRL levels, the HAL-induced increase in serum PRL concentrations was completely attenuated in the tail-vein bled animals (1.3-fold). In contrast, HAL produced a fivefold increase in serum PRL in animals where blood was collected under CO(2) anesthesia at euthanization. Hence, collection of blood from animals under CO(2) anesthesia at euthanization is an acceptable approach for detection of compounds that increase PRL. In summary, HAL-like compounds would be identified in the Tier I male and female battery primarily via increased serum PRL concentrations. RES-like compounds would be identified in the Tier I male battery via decreased gonadotropins and steroids and possibly in the Tier I female battery by a minimal increase in serum PRL concentrations. Compounds that produce a marginal increase in serum PRL when administered using single daily dosing can also be confirmed in an in vivo female battery with multiple dosing because this regimen increases the magnitude of the PRL increase. APO, a D(2) receptor agonist, was not detected in the in vivo male or female batteries, but in both instances the top dosage produced minimal decreases in body weight (99 to 96% of control). Hence, the proposed Tier I battery needs to be further evaluated with higher dosages of APO and other D(2) receptor agonists to determine whether it is capable of detecting such agents.


Assuntos
Apomorfina/farmacologia , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Sistema Endócrino/efeitos dos fármacos , Haloperidol/farmacologia , Reserpina/farmacologia , Androgênios/sangue , Animais , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sistema Endócrino/patologia , Estradiol/sangue , Feminino , Gonadotropinas Hipofisárias/sangue , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Hormônios Tireóideos/sangue , Testes de Toxicidade/métodos , Transformação Bacteriana/efeitos dos fármacos , Transformação Bacteriana/genética , Útero/efeitos dos fármacos , Útero/patologia
6.
Toxicol Sci ; 54(2): 338-54, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10774816

RESUMO

After previously examining 12 compounds with known endocrine activities, we have now evaluated 4 additional compounds in a Tier I screening battery for detecting endocrine-active compounds (EACs): a weak estrogen receptor (ER) agonist (coumestrol; COUM), an androgen receptor (AR) agonist (testosterone; TEST), a progesterone receptor (PR) agonist (progesterone; PROG), and a PR antagonist (mifepristone; RU486). The Tier I battery incorporates 2 short-term in vivo tests (5-day ovariectomized female battery; 15-day intact male battery) and an in vitro yeast transactivation system (YTS). The Tier I battery is designed to identify compounds that have the potential to act as agonists or antagonists to the estrogen, androgen, progesterone, or dopamine receptors; steroid biosynthesis inhibitors (aromatase, 5alpha-reductase, and testosterone biosynthesis); or compounds that alter thyroid function. In addition to the Tier I battery, a 15-day dietary restriction experiment was performed using male rats to assess confounding due to treatment-related decreases in body weight. In the Tier I female battery, TEST administration increased uterine weight, uterine stromal cell proliferation, and altered hormonal concentrations (increased serum testosterone [T] and prolactin [PRL]; and decreased serum FSH and LH). In the male battery, TEST increased accessory sex gland weights, altered hormonal concentrations (increased serum T, dihydrotestosterone [DHT], estradiol [E2], and PRL; decreased serum FSH and LH), and produced microscopic changes of the testis (Leydig cell atrophy and spermatid retention). In the YTS, TEST activated gene transcription in the yeast containing the AR or PR. In the female battery, COUM administration increased uterine weight, uterine stromal cell proliferation, and uterine epithelial cell height, and increased serum PRL concentrations. In the male battery, COUM altered hormonal concentrations (decreased serum T, DHT, E2; increased serum PRL) and, in the YTS, COUM activated gene transcription in the yeast containing the ER. In the female battery, PROG administration increased uterine weight, uterine stromal cell proliferation, and uterine epithelial cell height and altered hormonal concentrations (increased serum progesterone and decreased serum FSH and LH). In the male battery, PROG decreased epididymis and accessory sex gland weights, altered hormonal concentrations (decreased serum T, PRL, FSH, and LH; increased serum progesterone and E2), and produced microscopic changes of the testis (Leydig cell atrophy). In the YTS, PROG activated gene transcription in the yeast containing the AR or PR. In the female battery, RU486 administration increased uterine weight and decreased uterine stromal cell proliferation. In the male battery, RU486 decreased epididymis and accessory sex gland weights and increased serum FSH and LH concentrations. In the YTS, RU486 activated gene transcription in the yeast containing the ER, AR, or PR. Dietary restriction data demonstrate that confounding due to decrements in body weight are not observed when body weight decrements are 10% or less in the Tier I male battery. In addition, minimal confounding is observed at body decrements of 15% (relative liver weight, T3, and T4). Hence, compounds can be evaluated in this Tier I at levels that produce a 10% decrease in body weight without confounding of the selected endpoints. Using the responses obtained for all the endpoints in the Tier I battery, a distinct "fingerprint" was produced for each type of endocrine activity against which compounds with unknown activity can be compared. These data demonstrate that the described Tier I battery is useful for identifying EACs and they extend the compounds evaluated to 16.


Assuntos
Cumestrol/toxicidade , Sistema Endócrino/efeitos dos fármacos , Antagonistas de Hormônios/toxicidade , Mifepristona/toxicidade , Progesterona/toxicidade , Testosterona/toxicidade , Animais , Líquidos Corporais/efeitos dos fármacos , Líquidos Corporais/fisiologia , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Sistema Endócrino/patologia , Estro/efeitos dos fármacos , Feminino , Gônadas/efeitos dos fármacos , Gônadas/patologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Testes de Toxicidade/métodos , Útero/efeitos dos fármacos , Útero/patologia , Útero/fisiologia
7.
Toxicol Sci ; 51(1): 44-53, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10496676

RESUMO

In this report, p,p'-DDE, a weak androgen receptor (AR) antagonist, has been examined in a Tier I screening battery designed to detect endocrine-active compounds (EACs). The screening battery that was used to examine p,p'-DDE was an abbreviated version of a proposed Tier I screening battery (Cook et al., 1997, Regul. ToxicoL Pharmacol. 26, 60-68) that consisted of a 15-day intact male in vivo battery and an in vitro yeast transactivation system (YTS). In addition, strain sensitivity differences were evaluated using male Crl:CDIGS BR (CD) and Long-Evans (LE) rats. Finally, p,p'-DDE was examined in a Hershberger assay designed to detect AR agonists. In the in vivo male battery using CD rats, responses to p,p'-DDE included organ weight changes (increased relative liver weight and decreased absolute epididymis weight) and hormonal alterations (increased serum estradiol [E2] levels and decreased serum FSH and T4 levels). Responses to p,p'-DDE in LE rats included organ weight changes (increased relative liver weight, absolute epididymis weight, relative accessory sex gland [ASG] unit weight, as well as the individual component weights of the ASG [prostate and seminal vesicles]), and hormonal alterations (increased serum testosterone [T], E2, dihydrotestosterone [DHT], thyroid-stimulating hormone [TSH], and decreased T4 levels). These data demonstrate that there are considerable strain-sensitivity differences to p,p'-DDE exposure. The described in vivo male battery using CD rats did not identify p,p'-DDE as an EAC. In contrast, the in vivo male battery using LE rats identified p,p'-DDE as a EAC. Evaluation of the data for the LE rats demonstrate that p,p'-DDE appears to be acting as an AR antagonist whose primary effects are more potent centrally than peripherally. In the YTS for the AR, p,p'-DDE had an EC50 value of 3.5 x 10(-4) M; however, in the AR YTS competition assay, p,p'-DDE did not inhibit DHT binding to the AR. p,p'-DDE was inactive in the YTS containing the estrogen receptor or progesterone receptor at the concentrations evaluated. In the Hershberger assay, p,p'-DDE administration caused antiandrogen-like effects characterized by attenuation of the testosterone propionate-induced increases in reproductive-organ weights. In summary, these data suggest that strain selection will affect the ability to detect certain weak EACs. However, a Tier I screening battery consisting of both in vivo and in vitro endpoints would reduce the chance that weak-acting compounds such as p,p'-DDE would not be identified as potential EACs.


Assuntos
Diclorodifenil Dicloroetileno/toxicidade , Inseticidas/toxicidade , Testes de Toxicidade/métodos , Animais , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Flutamida/toxicidade , Hormônios Esteroides Gonadais/sangue , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Orquiectomia , Tamanho do Órgão/efeitos dos fármacos , Próstata/efeitos dos fármacos , Próstata/patologia , Ratos , Ratos Long-Evans , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Glândulas Seminais/efeitos dos fármacos , Glândulas Seminais/patologia , Testículo/efeitos dos fármacos , Testículo/patologia , Ativação Transcricional/efeitos dos fármacos
8.
Toxicol Sci ; 51(1): 54-70, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10496677

RESUMO

Phenobarbital (PB), a thyroid hormone excretion enhancer, and propylthiouracil (PTU), a thyroid hormone-synthesis inhibitor, have been examined in a Tier I screening battery for detecting endocrine-active compounds (EACs). The Tier I battery incorporates two short-term in vivo tests (5-day ovariectomized female battery and 15-day intact male battery using Sprague-Dawley rats) and an in vitro yeast transactivation system (YTS). In addition to the Tier I battery, thyroid endpoints (serum hormone concentrations, liver and thyroid weights, thyroid histology, and UDP-glucuronyltransferase [UDP-GT] and 5'-deiodinase activities) have been evaluated in a 15-day dietary restriction experiment. The purpose was to assess possible confounding of results due to treatment-related decreases in body weight. Finally, several thyroid-related endpoints (serum hormone concentrations, hepatic UDP-GT activity, thyroid weights, thyroid follicular cell proliferation, and histopathology of the thyroid gland) have been evaluated for their utility in detecting thyroid-modulating effects after 1, 2, or 4 weeks of treatment with PB or PTU. In the female battery, changes in thyroid endpoints following PB administration, were limited to decreased serum tri-iodothyronine (T3) and thyroxine (T4) concentrations. There were no changes in thyroid stimulating hormone (TSH) concentrations or in thyroid gland histology. In the male battery, PB administration increased serum TSH and decreased T3 and T4 concentrations. The most sensitive indicator of PB-induced thyroid effects in the male battery was thyroid histology (pale staining and/or depleted colloid). In the female battery, PTU administration produced increases in TSH concentrations, decreases in T3 and T4 concentrations, and microscopic changes (hypertrophy/hyperplasia, colloid depletion) in the thyroid gland. In the male battery, PTU administration caused thyroid gland hypertrophy/hyperplasia and colloid depletion, and the expected thyroid hormonal alterations (increased TSH, and decreased serum T3 and T4 concentrations). The dietary restriction study demonstrated that possible confounding of the data can occur with the thyroid endpoints when body weight decrements are 15% or greater. In the thyroid time course experiment, PB produced increased UDP-GT activity (at all time points), increased serum TSH (4-week time point), decreased serum T3 (1-and 2-week time points) and T4 (all time points), increased relative thyroid weight (2- and 4-week time points), and increased thyroid follicular cell proliferation (1- and 2-week time points). Histological effects in PB-treated rats were limited to mild colloid depletion at the 2- and 4-week time points. At all three time points, PTU increased relative thyroid weight, increased serum TSH, decreased serum T3 and T4, increased thyroid follicular cell proliferation, and produced thyroid gland hyperplasia/hypertrophy. Thyroid gland histopathology, coupled with decreased serum T4 concentrations, has been proposed as the most useful criteria for identifying thyroid toxicants. These data suggest that thyroid gland weight, coupled with thyroid hormone analyses and thyroid histology, are the most reliable endpoints for identifying thyroid gland toxicants in a short-duration screening battery. The data further suggest that 2 weeks is the optimal time point for identifying thyroid toxicants based on the 9 endpoints examined. Hence, the 2-week male battery currently being validated as part of this report should be an effective screen for detecting both potent and weak thyroid toxicants.


Assuntos
Antitireóideos/toxicidade , Fenobarbital/toxicidade , Propiltiouracila/toxicidade , Glândula Tireoide/efeitos dos fármacos , Animais , Líquidos Corporais/efeitos dos fármacos , Líquidos Corporais/metabolismo , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Estro/efeitos dos fármacos , Feminino , Glucuronosiltransferase/metabolismo , Gonadotropinas Hipofisárias/sangue , Iodeto Peroxidase/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Ovariectomia , Ratos , Ratos Sprague-Dawley , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Glândula Tireoide/patologia , Hormônios Tireóideos/sangue , Testes de Toxicidade/métodos , Útero/efeitos dos fármacos , Útero/metabolismo , Útero/patologia
9.
Drug Chem Toxicol ; 21(4): 417-47, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9839154

RESUMO

The chronic toxicity, oncogenicity, and mutagenicity of chlorotetrafluoroethane (HCFC-124) were evaluated. In the chronic toxicity/oncogenicity study, male and female rats were exposed to 0, 2000, 10,000, or 50,000 ppm HCFC-124 for 6 hr/day, 5 days/week, for 2 years. Body weights were obtained weekly during the first three months of the study and every other week for the remainder of the study. Food consumption was determined weekly. Clinical signs of toxicity were monitored throughout the study. An ophthalmological examination was performed on all animals prior to study start, and all surviving rats were examined at approximately 3, 12, and 24 months after study start. Clinical pathology was evaluated at 3, 6, 12, 18, and 24 months. An interim termination was conducted at 12 months. All surviving rats were necropsied at 24 months. A complete set of tissues was collected for microscopic examination, and selected tissues were weighed. There were no compound-related, adverse effects on body weight, food consumption, survival, clinical signs of toxicity, ophthalmoscopically observable ocular lesions, serum hormone concentrations, or clinical pathology parameters at any exposure concentration in either male or female rats. Compared to controls, urine fluoride was increased in males and females at all exposure concentrations, and plasma fluoride was increased in females at all exposure concentrations. Excretion of fluoride represents conversion of the parent molecule, and as such is not considered to be an adverse effect. There were no toxicologically significant, compound-related organ weight changes or gross or microscopic findings in male or female rats at any of the exposure concentrations tested. HCFC-124 was not toxic or carcinogenic in rats of either sex after inhalation exposure at concentrations of up to 50,000 ppm in this two-year chronic toxicity/oncogenicity study. After exposure to HCFC-124 for six hours per day, five days per week, for 24 months, the no-observed-adverse-effect level for male and female rats was 50,000 ppm. HCFC-124 was not mutagenic in Salmonella typhimurium strains TA1535, TA97, TA98, and TA100 with and without activation when evaluated at concentrations up to 60% HCFC-124 for 48 hours. No evidence of clastogenic activity was observed in cultured human lymphocytes at atmospheric concentrations up to 100% HCFC-124 for 3 hours, with and without metabolic activation. In vivo, no micronuclei were induced in mouse bone marrow cells following exposure of mice to concentrations of 99,000 ppm HCFC-124 6 hours/day for 2 days.


Assuntos
Clorofluorcarbonetos de Metano/toxicidade , Administração por Inalação , Animais , Peso Corporal/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Células Cultivadas , Etano Clorofluorcarbonos , Clorofluorcarbonetos de Metano/metabolismo , Aberrações Cromossômicas , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Fibroadenoma/epidemiologia , Fluoretos/sangue , Fluoretos/urina , Hormônios Esteroides Gonadais/sangue , Humanos , Leucócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Ratos , Salmonella typhimurium/efeitos dos fármacos , Taxa de Sobrevida , Fatores de Tempo , Testes de Toxicidade , Triglicerídeos/sangue
10.
Toxicol Sci ; 44(2): 116-42, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9742652

RESUMO

Over the past several years, there has been increasing concern that chemicals and pesticides found in the environment may mimic endogenous estrogens, potentially producing adverse effects in wildlife and human populations. Because estrogenicity is one of the primary concerns, a 90-day/one-generation reproduction study with 17 beta-estradiol was designed to set dose levels for future multigenerational reproduction and combined chronic toxicity/oncogenicity studies. The purpose of these studies is to evaluate the significance of a range of responses as well as to provide benchmark data for a risk assessment for chemicals with estrogen-like activities. This 90-day/one-generation reproduction study was conducted in male and female Crl:CD BR rats using dietary concentrations of 0, 0.05, 2.5, 10, and 50 ppm 17 beta-estradiol. Endpoints were chosen in order to evaluate both subchronic and reproductive toxicity. In addition, several mechanistic/biochemical endpoints were evaluated for their usefulness in follow-up studies. In the P1 generation, dietary administration of 2.5, 10, and 50 ppm 17 beta-estradiol produced dose-dependent decreases in body weight, body weight gain, food consumption, and food efficiency. At 10 and 50 ppm 17 beta-estradiol, minimal to mild nonregenerative anemia, lymphopenia, decreased serum cholesterol (50 ppm only), and altered splenic lymphocyte subtypes were also observed in the P1 generation. Additionally, at these concentrations, there were changes in the weights of several organs. Evidence of ovarian malfunction, characterized by reduced numbers of corpora lutea and large antral follicles, was observed at 2.5 ppm 17 beta-estradiol and above. Other pathologic changes in males and females fed 10 and 50 ppm 17 beta-estradiol included centrilobular hepatocellular hypertrophy; diffuse hyperplasia of the pituitary gland; feminization of the male mammary glands; mammary gland hyperplasia in females; increased number of cystic follicles in the ovary; hypertrophy of the endometrium and endometrial glands in the uterus; degeneration of seminiferous epithelium; and atrophy of the testes and the accessory sex glands. In the reproduction portion of this study, rats fed 10 or 50 ppm 17 beta-estradiol did not produce litters. While there was no evidence that the 50 ppm treated rats mated, 33.3% of the rats fed 10 ppm mated but did not produce litters. No effects on mating and fertility indices were observed in rats fed 0.05 and 2.5 ppm 17 beta-estradiol. Pup weights at birth were statistically decreased relative to control in the groups fed 0.05 and 2.5 ppm 17 beta-estradiol. Weights of the rats in the 0.05 ppm group recovered by postnatal day 4 and remained similar to control throughout the remainder of the study. The mean gestation length of the 0.05 ppm group was slightly, albeit not statistically significantly, shorter (0.5 days) than that of the control group, which may have contributed to the decrease in birth weight of the 0.05 ppm group. In contrast, the weights of the F1 generation rats fed 2.5 ppm 17 beta-estradiol remained decreased relative to the control group throughout the study. Parental administration of 17 beta-estradiol did not alter anogenital distance in male or female pups. The onset of sexual maturation, as measured by day of preputial separation in males and day of vaginal opening in females, was delayed in male rats fed 2.5 ppm (by 8.2 days) and was hastened in female rats fed 0.05 and 2.5 ppm (by 1.6 and 8.8 days, respectively). The age at vaginal opening ranged from 26 to 37, 26 to 35, and 21 to 25 days for rats fed 0, 0.05, and 2.5 ppm 17 beta-estradiol, respectively. Hence, the range of age at vaginal opening was similar between the control and 0.05 ppm group. The organ weight and pathologic alterations observed in the adult F1 generation rats were similar to those observed in the P1 generation rats. (ABSTRACT TRUNCATED)


Assuntos
Estradiol/toxicidade , Feto/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Animais , Feminino , Masculino , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Tamanho do Órgão/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/patologia , Gravidez , Ratos , Testículo/efeitos dos fármacos , Testículo/patologia , Vagina/efeitos dos fármacos , Vagina/patologia
11.
Toxicol Sci ; 44(2): 155-68, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9742654

RESUMO

A 90-day/one-generation reproduction study was conducted in male and female Crl:CD BR rats using dietary levels of 0, 0.05, 2.5, 10, and 50 ppm 17 beta-estradiol. The goals of this study were to set dose levels and evaluate several mechanistic endpoints for inclusion in multigeneration reproduction and combined chronic toxicity/oncogenicity studies with 17 beta-estradiol. In this report we discuss the effects of dietary 17 beta-estradiol exposure on serum hormonal levels and sperm parameters from P1 and F1 male rats. Sperm parameters were also evaluated in recovery P1 and F1 male rats that were fed control diets for 105 and 103 days, respectively, following 97 and 86-94 days of estradiol exposure, respectively. Measurement of Sertoli cell number from F1 male rats was performed to test the hypothesis that in utero exposure to estrogens will decrease Sertoli cell number and sperm production. Other findings from this 90-day/one-generation reproduction study are summarized elsewhere. 17 beta-Estradiol produced a dose-dependent decrease in body weight in P1 male rats at > or = 2.5 ppm and in the F1 male rats at 2.5 ppm. This decrease in body weight was due to a combination or reduced food consumption and food efficiency. In the recovery P1 males, body weight increased in the affected groups, albiet not to control levels, due to food consumption returning to control levels accompanied by an increase in food efficiency. However, in F1 males there was no corresponding rebound in body weight. In the P1 rats, exposure to 17 beta-estradiol decreased testis and epididymis weights in the 10 and 50 ppm groups, while no effects were seen in the P1 2.5 ppm group. In contrast, epididymis weights in the F1 and F1 recovery 2.5 ppm groups were statistically decreased; however, there were no histopathological effects observed. The decreases in testis weights in the P1 generation correlated with histopathologic evidence of interstitial cell atrophy and seminiferous tubule degeneration and reduced sperm production. Correlative changes in the epididymides of P1 rats were characterized by oligospermia or aspermia, the presence of germ cell debris in the lumen of tubules, and atrophy of epididymal tubules. 17 beta-Estradiol decreased testicular spermatid numbers, epididymal sperm numbers, and sperm motility in the P1 males in the 10 and 50 ppm groups, but not in the 2.5 ppm group. Following a 105-day recovery period in the P1 males, all sperm parameters and reproductive organ weights returned to control values except for the epididymal sperm count. Overall, the decline in testicular spermatid and epididymal sperm numbers in the P1 rats correlated with the reduced organ weights and the observed histopathological changes and appeared primarily related to the decrease in serum testosterone levels. In the F1 rats, no significant decreases were noted in the testicular spermatid number but a slight decrease in epididymal sperm number was seen in the 2.5 ppm group, which showed no evidence of recovery. Using morphometric analysis, no change was seen in the number of Sertoli cell nuclei per testis in F1 males. The pattern of hormonal responses seen in this study was characteristic of an estrogen receptor agonist such as 17 beta-estradiol: increased serum prolactin and decreased testosterone, luteinizing hormone, and follicle stimulating hormone levels. The data demonstrate that in utero and postnatal dietary administration of 17 beta-estradiol at levels which increased serum estradiol levels to approximately 400% of control and decreased testosterone levels to 33% of control did not reduce the number of Sertoli cell nuclei per testis.


Assuntos
Estradiol/toxicidade , Feto/efeitos dos fármacos , Hormônios/sangue , Testículo/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Masculino , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Prolactina/sangue , Ratos , Células de Sertoli/efeitos dos fármacos , Testículo/patologia , Testosterona/sangue
12.
Toxicol Sci ; 44(2): 169-84, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9742655

RESUMO

A Tier I screening battery for detecting endocrine active compounds (EACs) has been evaluated for its ability to identify 17 beta-estradiol, a pure estrogen receptor agonist. In addition, the responses obtained with the Tier I battery were compared to the responses obtained from F1 generation rats from a 90-day/one-generation reproduction study with 17 beta-estradiol in order to characterize the sensitivity of the Tier I battery against the sensitivity of an in utero exposure for detecting EACs. The Tier I battery incorporates two short-term in vivo tests (5-day ovariectomized female battery; 15-day intact male battery) and an in vitro yeast transactivation system (YTS) for identifying compounds that alter endocrine homeostasis. The Tier I female battery consists of traditional uterotrophic endpoints coupled with biochemical and hormonal endpoints. It is designed to identify compounds that are estrogenic/antiestrogenic or modulate dopamine levels. The Tier I male battery consists of organ weights coupled with microscopic evaluations and a comprehensive hormonal assessment. It is designed to identify compounds that have the potential to act as agonists or antagonists to the estrogen, androgen, progesterone, or dopamine receptor; steroid biosynthesis inhibitors (aromatase, 5 alpha-reductase, and testosterone biosynthesis); or compounds that alter thyroid function. The YTS is designed to identify compounds that bind to steroid hormone receptors (estrogen, androgen, and progesterone) and activate gene transcription. The profile generated for 17 beta-estradiol was characteristic of the responses expected with a pure estrogen receptor agonist. In the female battery, responses to 17 beta-estradiol included increases in uterine fluid imbibition, uterine weight, estrus conversion, uterine stromal cell proliferation, uterine epithelial cell height, uterine progesterone receptor content, serum prolactin and estradiol levels, and decreases in uterine estrogen receptor content and follicle stimulating hormone and luteinizing hormone levels. In the male battery, responses to 17 beta-estradiol included decreases in absolute testis and epididymides weights, decreases in relative weights for androgen-dependent tissues (prostate, seminal vesicles, and accessory sex gland unit), hormonal alterations (decreased serum testosterone, dihydrotestosterone, and LH and increased serum prolactin levels), and microscopic alterations of the testis and epididymides. In the YTS for the estrogen receptor, 17 beta-estradiol had an EC50 value of 7.2 x 10(-9) M, while DHT and progesterone had little cross-activation. The androgen and progesterone receptor systems were less selective in that 17 beta-estradiol activated these systems within 3 orders of magnitude of the primary ligand. In the 90-day/one-generation reproduction study, responses to dietary administration of 17 beta-estradiol included alterations in organ weights, developmental landmarks, and hormonal levels. Comparison of the responses obtained with our Tier I battery and an in utero exposure demonstrates that the Tier I screening battery is as sensitive as an in utero exposure for detecting 17 beta-estradiol-induced alterations in hormonal homeostasis.


Assuntos
Estradiol/toxicidade , Feto/efeitos dos fármacos , Receptores de Estrogênio/agonistas , Animais , Feminino , Hormônios/sangue , Masculino , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Ratos , Receptores de Estrogênio/análise , Receptores de Progesterona/análise , Saccharomyces cerevisiae/efeitos dos fármacos , Sensibilidade e Especificidade , Testículo/efeitos dos fármacos , Útero/efeitos dos fármacos
13.
Toxicol Sci ; 46(2): 235-46, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10048126

RESUMO

Physiologically based pharmacokinetic (PBPK) models require estimates of catalytic rate constants controlling the metabolism of xenobiotics. Usually, these constants are derived from whole tissue homogenates wherein cellular architecture and enzyme compartmentation are destroyed. Since the nasal cavity epithelium is composed of a heterogeneous cell population measurement of xenobiotic metabolizing enzymes using homogenates could yield artifactual results. In this article a method for measuring rates of metabolism of vinyl acetate, a metabolism-dependent carcinogen, is presented that uses whole-tissue samples and PBPK modeling techniques to estimate metabolic kinetic parameters in tissue compartments. The kinetic parameter estimates were compared to those derived from homogenate experiments using two methods of tissue normalization. When the in vitro gas uptake constants were compared to homogenate-derived values, using a normalization procedure that does not account for tissue architecture, there was poor agreement. Homogenate-derived values from rat nasal tissue were 3- to 23-fold higher than those derived using the in vitro gas uptake method. When the normalization procedure for the rat homogenate-derived values took into account tissue architecture, a good agreement was observed. Carboxylesterase activity in homogenates of human nasal tissues was undetectable. Using the in vitro gas uptake technique, however, carboxylesterase activity was detected. Rat respiratory carboxylesterase and aldehyde dehydrogenase activities were about three and two times higher than those of humans, respectively. Activities of the rat olfactory enzymes were about equivalent to those of humans. K(m) values did not differ between species. The results suggest that the in vitro gas uptake technique is useful for deriving enzyme kinetic constants where effects of tissue architecture are preserved. Furthermore, the results suggest that caution should be exercised when scaling homogenate-derived values to whole-organ estimates, especially in organs of cellular heterogeneity.


Assuntos
Aldeído Desidrogenase/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Gases/farmacocinética , Mucosa Nasal/metabolismo , Compostos de Vinila/metabolismo , Idoso , Animais , Carboxilesterase , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Ratos , Especificidade da Espécie , Estatística como Assunto
14.
Toxicol Sci ; 46(1): 45-60, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9928668

RESUMO

After previously examining an estrogen receptor agonist (17beta-estradiol), several additional compounds have been evaluated in a Tier I screening battery for detecting endocrine-active compounds (EACs): an estrogen receptor antagonist (ICI-182,780, ICI), an androgen receptor antagonist (flutamide, FLUT), a testosterone biosynthesis inhibitor (ketoconazole, KETO), a 5alpha-reductase inhibitor (finasteride, FIN), and an aromatase inhibitor (anastrozole, ANA). The Tier I battery incorporates two short-term in vivo tests (a 5-day ovariectomized female battery and a 15-day intact male battery) and an in vitro yeast transactivation system (YTS). The Tier I battery is designed to identify compounds that have the potential to act as agonists or antagonists to the estrogen, androgen, progesterone, or dopamine receptors, steroid biosynthesis inhibitors (aromatase, 5alpha-reductase, and testosterone biosynthesis), or compounds that alter thyroid function. ICI administration decreased uterine estrogen and progesterone receptor number in the female battery, increased serum follicle-stimulating hormone (FSH) levels and caused spermatid retention in the male battery, and activated gene transcription in the YTS containing the estrogen receptor. FLUT administration increased uterine stromal cell proliferation in the female battery and decreased weights for all androgen-dependent tissues, induced Leydig cell hyperplasia, and caused hormonal alterations (increased testosterone (T), estradiol (E2), dihydrotestosterone (DHT), luteinizing hormone (LH), and FSH) in the male battery, and competed for binding to the androgen receptor in the YTS competition assay. In the male battery KETO decreased weights for all androgen-dependent tissues, caused hormonal alterations (decreased T and DHT and increased LH and FSH), and induced spermatid retention. FIN decreased seminal vesicle and accessory sex gland (ASG) unit weight and caused hormonal alterations (decreased DHT and increased LH, and PRL) in the male battery. KETO was judged not to affect any of the endpoints in the female battery. ANA decreased ASG unit weight and serum E2 levels in the male battery. Using the responses obtained for all the endpoints in the Tier I battery, a distinct "fingerprint" was produced for each type of endocrine activity against which compounds with unknown activity can be compared. These data demonstrate that the described Tier I battery is useful for identifying EACs.


Assuntos
Sistema Endócrino/efeitos dos fármacos , Testes de Toxicidade/métodos , Animais , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Sistema Endócrino/patologia , Células Epiteliais/efeitos dos fármacos , Estro/efeitos dos fármacos , Feminino , Hormônios Esteroides Gonadais/sangue , Hormônios/sangue , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Reprodutibilidade dos Testes , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Fatores Sexuais , Hormônios Tireóideos/sangue , Útero/citologia , Útero/efeitos dos fármacos , Útero/metabolismo
15.
Mutat Res ; 380(1-2): 155-65, 1997 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-9385396

RESUMO

Hexamethylphosphoramide (HMPA) is a rat nasal carcinogen that induces squamous cell carcinomas in the anterior portions of the nasal cavity following chronic inhalation exposures as low as 50 ppb. These tumors may arise as a result of P-450-mediated release of formaldehyde (HCHO), a known rat nasal carcinogen. The goal of this research was to investigate early responses of the nasal epithelium to inhaled HMPA. Rats were exposed nose-only to approximately 3 ppm HMPA for 6 h, and killed 18, 48, 96 or 144 h post-exposure. In a separate study, rats were exposed nose-only for 6 h for 1, 2, 3, or 5 consecutive days and killed 18 or 96 h post-exposure. With both single and repeated doses of HMPA, there was no evidence of cytotoxicity in the anterior nose. Olfactory degeneration and necrosis of the dorsal meatus, Bowman's glands and tips of the ethmoid turbinates increased in severity with repeated exposures to HMPA. Cell proliferation was assessed in levels of nasal tissue that included regions of squamous, respiratory, transitional and olfactory epithelium. Regional induction of cell proliferation was measured by BrdU incorporation, and reported as the number of labeled cells/mm basement membrane. At 18 h after a single exposure, there was an increase in cell proliferation in squamous epithelium, which returned to control levels within 48 h. A transitory increase in cell proliferation was observed regions of respiratory and transitional epithelium, although the response of each tissue, in terms of magnitude and peak time of response post-exposure, also differed. Along the dorsal meatus in Level 9, olfactory labeling initially decreased, returned to control levels by 96 h, but again declined at 144 h post-exposure. In repeat dose studies, the squamous epithelium response was variable 18 h post-exposure. For respiratory and transitional epithelium, increased cell proliferation 18 h post-exposure was correlated with increased dose (exposure) of HMPA. Cell proliferation responses following two or more exposures returned to near control levels within 96 h post-exposure. In conclusion, HMPA induced cell proliferation, but not cytotoxicity, in the anterior nose at approximately 3 ppm. These data suggest that HMPA induces proliferative, perhaps mitogenic, responses in the nasal epithelium, and this response may facilitate the fixation of low level genetic damage induced by liberated HCHO.


Assuntos
Carcinógenos/toxicidade , Hempa/toxicidade , Cavidade Nasal/efeitos dos fármacos , Administração por Inalação , Animais , Divisão Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Hempa/administração & dosagem , Masculino , Ratos , Fatores de Tempo
16.
Environ Health Perspect ; 105 Suppl 5: 1219-22, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9400727

RESUMO

This study compared pulmonary effects of para-aramid respirable-sized, fiber-shaped particles (RFP) (p-aramid fibrils) and chrysotile asbestos fiber exposures in rats. Additional p-aramid inhalation studies were conducted in hamsters to compare species responses. The hamster results are preliminary. The parameters studied were clearance/biopersistence of inhaled p-aramid RFP or size-separated asbestos fibers as well as pulmonary cell proliferation and inflammation indices after 2-week inhalation exposures. Rats were exposed nose only to chrysotile asbestos fibers at concentrations of 459 and 782 fibers/ml or to p-aramid RFP at 419 or 772 fibrils/ml. Hamsters were exposed whole body to p-aramid RFP at concentrations of 358 and 659 fibrils/ml. Subsequently, animals were assessed immediately (time 0) as well as 5 days (10 days for hamsters), 1, 3, 6, and 12 months postexposure. Lung burdens for the p-aramid-exposed rats were 4.8 x 10(7) and 7.6 x 10(7) fibrils/lung, with similar numbers of chrysotile fibers > 5 microns recovered from the lungs of asbestos-exposed rats. In comparison, 1.4 x 10(6) fibrils/lung were recovered in the high-dose hamster group. Biopersistence studies in p-aramid-exposed rats and hamsters demonstrated an initial increase (relative to time 0) in retained p-aramid fibrils during the first month postexposure, which indicated breakage or shortening of inhaled fibrils. This result was associated with a progressive reduction, and increased residence time in the lung, in the mean lengths of the fibrils, which signified biodegradability of inhaled p-aramid fibrils in both species. In contrast, clearance of short chrysotile asbestos fibers was rapid, but clearance of the long chrysotile fibers was slow or insignificant, as evidenced by a progressive increase over time in the mean lengths of fibers recovered from the lungs of exposed rats. Two-week, high-dose exposures to p-aramid in both rats and hamsters produced transient increases in pulmonary inflammatory and cell proliferative responses. In contrast, inhalation of size-separated chrysotile asbestos fibers in rats produced persistent increases in cell labeling indices of airway, alveolar, and subpleural cells measured through a period of 1 to 3 months postexposure. These results suggest that inhaled p-aramid RFP are biodegradable in the lungs of exposed rats and hamsters. In contrast, exposures to chrysotile asbestos fibers in rats resulted in a selective pulmonary retention of long chrysotile fibers.


Assuntos
Asbestos Serpentinas/farmacocinética , Asbestos Serpentinas/toxicidade , Carcinógenos/farmacocinética , Carcinógenos/toxicidade , Pulmão/metabolismo , Pulmão/patologia , Polímeros/farmacocinética , Polímeros/toxicidade , Administração por Inalação , Animais , Antimetabólitos/toxicidade , Asbestos Serpentinas/administração & dosagem , Biodegradação Ambiental , Bromodesoxiuridina/toxicidade , Carcinógenos/administração & dosagem , Divisão Celular/efeitos dos fármacos , Cricetinae , Masculino , Mesocricetus , Polímeros/administração & dosagem , Ratos , Ratos Endogâmicos
17.
Toxicol Appl Pharmacol ; 146(1): 116-26, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9299603

RESUMO

A8947 is a member of the sulfonyl urea class of compounds and is the active ingredient in a commercial broad leaf herbicide. This compound has been shown to produce pancreatic hypertrophy in rats, mice, and dogs. The objectives of this study were to investigate the mechanism(s) for the A8947 induction of pancreatic acinar cell hypertrophy and proliferation and to evaluate whether these pancreatic changes are reversible. A8947 was fed to male Crl:CD BR rats for up to 28 days (0, 300, 10,000, 30,000 ppm) or 56 days (0, 30,000 ppm). Rats were terminated on Test Days 7, 14, and 28 to assess the time course and dose response for the A8947-induced pancreatic changes, while rats terminated on Test Day 56 were used to assesss the reversibility of the pancreas effects at 30,000 ppm A8947. A8947 produced significant increases in pancreatic weight and acinar cell proliferation and diffuse acinar cell hypertrophy in 7 days at 10,000 and 30,000 ppm dose levels. By Day 14, absolute pancreas weights in the 10,000 and 30,000 ppm groups were maximally increased and remained at these levels throughout the study. In contrast, acinar cell proliferation in the 30,000 ppm group was still elevated at Test Day 14, but attenuated relative to the 7-day response, and returned to control levels by Test Day 28. No effects were observed at 300 ppm after a 28-day exposure period, while complete reversibility of A8947-induced pancreatic effects was demonstrated at 30,000 ppm following a 1-month recovery period (Test Day 56). Cholecystokinin (CCK) levels were increased by A8947 and closely followed the time course for pancreatic changes. MK-329, a specific CCKA receptor antagonist, completely ablated the ability of 30,000 ppm A8947 to increase pancreas weight following 7 days of exposure. A8947 did not bind the CCKA receptor in a receptor competition assay, negating any potential agonist mechanism. A8947 did, however, inhibit trypsin in vitro, suggesting a mechanism of action similar to that of raw soy protein, in which trypsin inhibition in vivo results in increased CCK levels followed by pancreatic acinar cell hypertrophy and proliferation.


Assuntos
Herbicidas/toxicidade , Pâncreas/efeitos dos fármacos , Pirimidinas/toxicidade , Inibidores da Tripsina/toxicidade , Animais , Benzodiazepinonas/farmacologia , Colecistocinina/sangue , Devazepida , Relação Dose-Resposta a Droga , Hipertrofia , Masculino , Pâncreas/patologia , Ratos , Receptor de Colecistocinina A , Receptores da Colecistocinina/agonistas
18.
Toxicol Appl Pharmacol ; 145(2): 425-36, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9266817

RESUMO

Several peroxisome proliferators have been shown to produce pancreatic acinar cell hyperplasia/adenocarcinomas in 2-year bioassays with rats: ammonium perfluorooctanoate (C8), clofibrate, methylclofenapate, HCFC-123, and Wyeth-14,643 (WY). We have used in vitro (C8, WY) and in vivo (WY) approaches to examine several possible mechanisms of pancreatic tumorigenesis by peroxisome proliferating compounds. These mechanisms include cholecystokinin receptor agonism (CCK(A)), trypsin inhibition, alterations in gut fat content, cholestasis, and altered bile flow/composition. All of these mechanisms enhance pancreatic growth either by binding to the CCK(A) receptor or by increasing plasma CCK levels. In vitro experiments using a receptor competition binding assay demonstrated that WY and C8 do not bind directly to the CCK(A) receptor. In a continuous spectrophotometric assay, WY and C8 also failed to inhibit trypsin, a common mechanism for increasing plasma CCK levels. These in vitro results suggested that WY was not acting via the two most common mechanisms for modulation of pancreas growth. Two types of in vivo experiments were conducted. The subchronic study (2-month duration) was designed primarily to detect early changes in pancreatic growth such as those mediated by compounds that inhibit trypsin or act as CCK(A) receptor agonists. The chronic study (6 months) was designed primarily to evaluate whether the pancreatic lesions were secondary to hepatic changes such as cholestasis and/or altered bile flow/composition. In the in vivo experiments, male Crl:CDBR rats were fed diets containing 0 or 100 ppm WY. In the subchronic study WY-treated rats had a twofold increase in mean relative liver weights, an eightfold increase in hepatic peroxisomal proliferation, and a fourfold increase in hepatocyte cell proliferation after 1 week which remained elevated throughout the 2 months of treatment. In contrast, no pancreatic weight effects, increases in plasma CCK, or acinar cell proliferation was seen through 2 months in the WY group when compared to the control group. Fecal fat concentrations were also measured at 2 months and demonstrated no difference between control and WY-treated animals. The absence of any early pancreas changes in the subchronic study is consistent with the in vitro data which demonstrated that WY is not a CCK(A) agonist or a trypsin inhibitor. The chronic study demonstrated increases in pancreatic weights at 3 months (6% above control) and 6 months (17% above control), as well as increased CCK plasma levels in the WY-treated group. Liver effects in the chronic study paralleled those of the subchronic time points. Clinical pathology endpoints including increased serum concentrations of bile acids, alkaline phosphatase, and bilirubin were indicative of cholestasis in the chronic WY-treated group. The cholestasis may be responsible for the downward trend in total bile acid output, both of which may contribute to the modest increases in plasma CCK levels. These results indicate that chronic exposure to WY causes liver alterations such as cholestasis, which may increase plasma concentrations of CCK. Hence, WY may induce pancreatic acinar cell adenomas/adenocarcinomas via a mild but sustained increase in CCK levels secondary to hepatic cholestasis.


Assuntos
Carcinógenos/toxicidade , Pâncreas/efeitos dos fármacos , Neoplasias Pancreáticas/induzido quimicamente , Pirimidinas/toxicidade , Adenoma/induzido quimicamente , Animais , Ligação Competitiva/efeitos dos fármacos , Carcinoma de Células Acinares/induzido quimicamente , Colecistocinina/agonistas , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Microcorpos/efeitos dos fármacos , Mutagênicos/toxicidade , Ratos , Ratos Endogâmicos , Receptores da Colecistocinina/efeitos dos fármacos , Inibidores da Tripsina/toxicidade
19.
Food Chem Toxicol ; 35(3-4): 393-407, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9207902

RESUMO

Male and female Fischer 344 rats were exposed to methyl methacrylate (MMA) monomer vapours at 0, 25, 100 and 400 ppm, 6 hr/day, 5 days/wk for 24 months and male and female Golden hamsters were exposed to similar vapour concentrations of MMA for 18 months. Parameters monitored throughout the study included clinical signs, individual body weights, haematology, clinical chemistry (rats only) and urinalyses (rats only). 10 rats per sex per exposure group were killed after 13 and 52 wk of exposure and all surviving rats were killed during wk 104-106. All surviving hamsters were killed at wk 78. Mortality and haematological, clinical chemistry and urinalyses parameters were not affected by MMA exposure. Body weights of male rats were not affected by exposure to MMA while body weights of female rats exposed to 400 ppm were lower than control values after wk 52. Male and female hamsters exposed to 400 ppm had body weight decreases ranging from 9 to 12% after wk 48. The nasal cavity was identified as the target organ for chronic toxicity in male and female rats exposed to 100 or 400 ppm. The microscopic nasal cavity changes occurred primarily in olfactory epithelium lining the dorsal meatus and consisted of degeneration of neuroepithelium, basal cell hyperplasia and atrophy of Bowman's glands. Hamsters did not have demonstrable nasal cavity microscopic changes. Chronic exposure to MMA vapour did not cause tumours in either rats or hamsters.


Assuntos
Metilmetacrilatos/toxicidade , Cavidade Nasal/efeitos dos fármacos , Administração por Inalação , Animais , Peso Corporal/efeitos dos fármacos , Testes de Carcinogenicidade , Cricetinae , Epitélio/efeitos dos fármacos , Epitélio/patologia , Feminino , Masculino , Mesocricetus , Metilmetacrilatos/administração & dosagem , Cavidade Nasal/citologia , Cavidade Nasal/patologia , Neoplasias/induzido quimicamente , Neoplasias/patologia , Ratos , Ratos Endogâmicos F344 , Taxa de Sobrevida
20.
Drug Chem Toxicol ; 20(1-2): 1-10, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9183558

RESUMO

A two-year mechanistic bioassay in male Crl:CD BR rats was initiated with 50 ppm Wyeth-14,643 (WY) to investigate the relationship between peroxisome proliferating compounds and Leydig cell adenoma formation. After 154 days, the survival rate in the WY group decreased below control levels. After 300 days, the dose was lowered to 25 ppm for the remainder of the study. Gross examination of WY-treated rats either found dead or euthanized in extremis revealed hemorrhages at several sites. To investigate this observation, blood was then collected on test day 281 from 10 randomly selected control and WY-treated rats and a clinical pathological examination was performed. The WY-treated rats had significantly decreased red blood cell count, hemoglobin, and hematocrit, and elevated platelet counts. In the WY-treated rats, prothrombin times in undiluted plasma were similar to the controls, but were markedly prolonged in 2 of 10 rats when the plasma samples were diluted to 25%. Subsequently, blood was collected prior to sacrificing WY-treated rats which were exhibiting clinical signs of anemia. These rats had prolonged prothrombin times, activated partial thromboplastin time, and thrombin clot time when compared to laboratory historical control data (116.7 vs 13.3, 116.4 vs 13.7, and 42.4 vs 25.7 seconds, respectively). In a subsequent, ongoing study, Vitamin K was added to control and WY-treated diets (100 ppm). No survival differences between control and WY-treated rats occurred through 260 days in this second study. These new data suggest that deaths in the WY-treated group in our initial study were due to a vitamin K deficiency. The role of increased serum estradiol, its effects on blood coagulation, and enhanced hepatic cell proliferation in the vitamin K-dependent coagulation processes warrant further investigation.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Carcinógenos/toxicidade , Tumor de Células de Leydig/induzido quimicamente , Mutagênicos/toxicidade , Pirimidinas/toxicidade , Neoplasias Testiculares/induzido quimicamente , Análise de Variância , Animais , Bioensaio , Coleta de Amostras Sanguíneas , Relação Dose-Resposta a Droga , Contagem de Eritrócitos/efeitos dos fármacos , Hematócrito , Hemoglobinas/metabolismo , Tumor de Células de Leydig/sangue , Tumor de Células de Leydig/mortalidade , Masculino , Microcorpos/efeitos dos fármacos , Tempo de Tromboplastina Parcial , Contagem de Plaquetas/efeitos dos fármacos , Tempo de Protrombina , Pirimidinas/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Neoplasias Testiculares/sangue , Neoplasias Testiculares/mortalidade , Trombina/metabolismo , Deficiência de Vitamina K/sangue , Deficiência de Vitamina K/induzido quimicamente , Deficiência de Vitamina K/mortalidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...