Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Crit Rev Toxicol ; 46(9): 785-833, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27347635

RESUMEN

The US Environmental Protection Agency Endocrine Disruptor Screening Program (EDSP) is a tiered screening approach to determine the potential for a chemical to interact with estrogen, androgen, or thyroid hormone systems and/or perturb steroidogenesis. Use of high-throughput screening (HTS) to predict hazard and exposure is shifting the EDSP approach to (1) prioritization of chemicals for further screening; and (2) targeted use of EDSP Tier 1 assays to inform specific data needs. In this work, toxicology data for three triazole fungicides (triadimefon, propiconazole, and myclobutanil) were evaluated, including HTS results, EDSP Tier 1 screening (and other scientifically relevant information), and EPA guideline mammalian toxicology study data. The endocrine-related bioactivity predictions from HTS and information that satisfied the EDSP Tier 1 requirements were qualitatively concordant. Current limitations in the available HTS battery for thyroid and steroidogenesis pathways were mitigated by inclusion of guideline toxicology studies in this analysis. Similar margins (3-5 orders of magnitude) were observed between HTS-predicted human bioactivity and exposure values and between in vivo mammalian bioactivity and EPA chronic human exposure estimates for these products' registered uses. Combined HTS hazard and human exposure predictions suggest low priority for higher-tiered endocrine testing of these triazoles. Comparison with the mammalian toxicology database indicated that this HTS-based prioritization would have been protective for any potential in vivo effects that form the basis of current risk assessment for these chemicals. This example demonstrates an effective, human health protective roadmap for EDSP evaluation of pesticide active ingredients via prioritization using HTS and guideline toxicology information.


Asunto(s)
Disruptores Endocrinos/toxicidad , Fungicidas Industriales/toxicidad , Pruebas de Toxicidad/métodos , Triazoles/toxicidad , Bioensayo , Disruptores Endocrinos/clasificación , Disruptores Endocrinos/normas , Fungicidas Industriales/clasificación , Fungicidas Industriales/normas , Nitrilos/toxicidad , Triazoles/clasificación , Triazoles/normas , Estados Unidos
2.
Regul Toxicol Pharmacol ; 75: 89-104, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26472101

RESUMEN

A strategic and comprehensive program in which toxicokinetic (TK) measurements are made for all agrochemicals undergoing toxicity testing (both new compounds and compounds already registered for use) is described. This approach provides the data to more accurately assess the toxicokinetics of agrochemicals and their metabolites in laboratory animals and humans. Having this knowledge provides the ability to conduct more insightful toxicity studies, refine and interpret exposure assessments and reduce uncertainty in risk assessments. By developing a better understanding of TK across species, including humans via in vitro metabolism studies, any differences across species in TK can be identified early and the most relevant species can be selected for toxicity tests. It also provides the ability to identify any non-linearities in TK as a function of dose, which in turn can be used to identify a kinetically derived maximum dose (KMD) and avoid dosing inappropriately outside of the kinetic linear range. Measuring TK in key life stages also helps to identify changes in ADME parameters from in utero to adults. A robust TK database can also be used to set internal concentration based "Reference Concentrations" and Biomonitoring Equivalents (BE), and support selection of Chemical Specific Adjustment Factors (CSAF). All of these factors support the reduction of uncertainty throughout the entire risk assessment process. This paper outlines how a TK research strategy can be integrated into new agrochemical toxicity testing programs, together with a proposed Framework for future use.


Asunto(s)
Agroquímicos/farmacocinética , Agroquímicos/toxicidad , Medición de Riesgo/métodos , Animales , Relación Dosis-Respuesta a Droga , Exposición a Riesgos Ambientales/análisis , Humanos , Toxicocinética
3.
Regul Toxicol Pharmacol ; 82: 127-139, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27663666

RESUMEN

The European Centre for the Ecotoxicology and Toxicology of Chemicals (ECETOC) organised a workshop to discuss the state-of-the-art research on noncoding RNAs (ncRNAs) as biomarkers in regulatory toxicology and as analytical and therapeutic agents. There was agreement that ncRNA expression profiling data requires careful evaluation to determine the utility of specific ncRNAs as biomarkers. To advance the use of ncRNA in regulatory toxicology, the following research priorities were identified: (1) Conduct comprehensive literature reviews to identify possibly suitable ncRNAs and areas of toxicology where ncRNA expression profiling could address prevailing scientific deficiencies. (2) Develop consensus on how to conduct ncRNA expression profiling in a toxicological context. (3) Conduct experimental projects, including, e.g., rat (90-day) oral toxicity studies, to evaluate the toxicological relevance of the expression profiles of selected ncRNAs. Thereby, physiological ncRNA expression profiles should be established, including the biological variability of healthy individuals. To substantiate the relevance of key ncRNAs for cell homeostasis or pathogenesis, molecular events should be dose-dependently linked with substance-induced apical effects. Applying a holistic approach, knowledge on ncRNAs, 'omics and epigenetics technologies should be integrated into adverse outcome pathways to improve the understanding of the functional roles of ncRNAs within a regulatory context.


Asunto(s)
ARN no Traducido/genética , Pruebas de Toxicidad/métodos , Toxicología/métodos , Animales , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Genómica , Humanos , Modelos Animales , ARN no Traducido/metabolismo , Reproducibilidad de los Resultados , Factores de Tiempo
4.
Artículo en Inglés | MEDLINE | ID: mdl-25652268

RESUMEN

Histiotrophic nutrition via the visceral yolk sac is an essential nutritional pathway of the rodent conceptus, and inhibition of this pathway may cause growth retardation, malformations, and death in rodent embryos. Morphologic differences among species during early development indicate that the visceral yolk sac histiotrophic nutrition pathway may be of lesser importance in nonrodent species, including humans. Here, comparative studies were conducted with inhibitors of different steps in the visceral yolk sac histiotrophic nutrition pathway to determine whether the rabbit is similarly responsive to the rat. Early somite stage New Zealand White rabbit and Crl:CD(SD) rat conceptuses (gestation day 9, rabbits; gestation day 10, rats) were exposed for 48 hr to three different histiotrophic nutrition pathway inhibitors using whole embryo culture techniques, after which they were evaluated for growth and malformations. Cubilin antibody, an inhibitor of endocytosis, reduced growth and development and increased malformations in both rat and rabbit embryos, although the rabbit appeared more sensitive. Leupeptin, a lysosomal cysteine protease inhibitor, also impaired growth and development and increased malformations in rat embryos, while in the rabbit it induced malformations and a slight decrease in morphology score but had no effect upon growth. Trypan blue, an inhibitor of endocytosis and endosome maturation, affected all measures in both species to a similar degree at the highest concentration (2500 µg/ml), but rat embryos responded to a greater extent at lower concentrations. Although the specific adverse outcomes appear to be different, these results demonstrate that rabbits, like rats, are sensitive to inhibitors of the histiotrophic nutrition pathway.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales/efectos de los fármacos , Feto/efectos de los fármacos , Feto/fisiología , Animales , Anticuerpos/farmacología , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Femenino , Leupeptinas/farmacología , Conejos , Ratas Sprague-Dawley , Receptores de Superficie Celular/inmunología , Coloración y Etiquetado , Azul de Tripano/farmacología
5.
Regul Toxicol Pharmacol ; 72(2): 405-22, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25846366

RESUMEN

Based on the exposure potential to humans and environment, pronamide was one of 52 chemicals on the first list evaluated under US EPA's Endocrine Disruptor Screening Program (EDSP). The purpose of EDSP is to screen chemicals for their potential to interact with estrogen-, androgen-, or thyroid-signaling pathways. A battery of 11 Tier 1 assays was completed for pronamide in accordance with EDSP test guidelines. In addition, Other Scientifically Relevant Information, which included existing data from regulatory guideline studies and published literature, was used in a weight-of-evidence (WoE) evaluation of potential endocrine activity. The WoE conclusion is that pronamide does not interact directly with estrogen, androgen, or thyroid receptors or post-receptor events. Across in vivo studies, the liver is consistently and reproducibly the target organ for pronamide's effects. Pronamide activates hepatocytic nuclear receptors (including constitutive androstane receptor), induces hepatic enzymes, produces hepatocellular hypertrophy and increases liver weights. These changes are coupled with increased metabolic activity and a subsequent increased metabolism and/or clearance of both steroid and thyroid hormones. Thus, while pronamide alters some endocrine-sensitive endpoints in EDSP Tier 1 assays, effects on liver metabolism likely explain altered hormone levels and indirect endocrine changes.


Asunto(s)
Benzamidas/toxicidad , Disruptores Endocrinos/toxicidad , Herbicidas/toxicidad , Hígado/efectos de los fármacos , Animales , Hormonas Gonadales/metabolismo , Gonadotropinas Hipofisarias/metabolismo , Humanos , Hígado/metabolismo , Hormonas Tiroideas/metabolismo
6.
Regul Toxicol Pharmacol ; 71(3): 541-51, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25707858

RESUMEN

Pronamide, a selective, systemic, pre- and post-emergence herbicide, caused an increased incidence of thyroid follicular cell adenomas in a rat carcinogenicity study. Thyroid tumors, as well as liver and pituitary changes, were limited only to the high-dose group. The evidence for and against specific potential modes of action (MoAs) for rat thyroid follicular cell adenomas and their relevance to humans is discussed. Pronamide is not mutagenic and therefore, direct DNA reactivity is not relevant as a MoA. The hypothesized MoA for this effect is altered homeostasis of the hypothalamic-pituitary-thyroid (HPT) axis mediated by the induction of hepatic enzymes, including uridine diphosphate glucuronosyltransferase (UGT). Evaluation of data from a series of regulatory guideline and MoA studies aimed at identifying the causative and associated key events supported a UGT-mediated MoA in the development of thyroid follicular tumors. This MoA for pronamide-induced thyroid tumors in rats, which involves increased thyroid hormone metabolism/clearance, altered thyroid hormone homeostasis and HPT stimulation is not considered relevant to humans based on quantitative species differences, making rats markedly more sensitive than humans to thyroid perturbations.


Asunto(s)
Adenoma/inducido químicamente , Benzamidas/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Herbicidas/toxicidad , Neoplasias de la Tiroides/inducido químicamente , Adenoma/metabolismo , Adenoma/patología , Animales , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Glucuronosiltransferasa/metabolismo , Humanos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Ratas , Medición de Riesgo , Especificidad de la Especie , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Hormonas Tiroideas/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Regulación hacia Arriba
7.
Regul Toxicol Pharmacol ; 72(2): 394-404, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25846364

RESUMEN

Dietary exposure to pronamide resulted in higher incidences of Leydig cell tumors (LCT) at 1000ppm in a 2-year cancer bioassay, but there were no testes effects at 40 or 200ppm, and no testes effects at 12-months at any concentration. A 90-day mode-of-action (MoA) study was conducted at concentrations of 0, 200, 1000 and 2000ppm. Standard parameters and stereological and proliferation analyses of LCs, targeted testis and liver gene expression, in vitro metabolism of testosterone by liver microsomes, and quantification of serum hormones and testosterone metabolites were evaluated. Increased testosterone metabolism due to increases in hepatic microsomal activity, alterations in serum hormone levels, and other data suggest that LCTs were mediated through a perturbation of the HPG-axis. Data suggest that this occurs after a threshold of exposure is reached, indicating a nonlinear/threshold dose-response. Pronamide-induced rat LCTs mediated by alterations to the HPG-axis have low relevance to humans due to quantitative differences in sensitivity between rats and humans to LCTs. Pronamide displayed no genotoxicity or direct endocrine effects. A margin of exposure approach for risk assessment and derivation of the chronic reference dose based on a point of departure of 200ppm is most appropriate and protective of human health.


Asunto(s)
Benzamidas/toxicidad , Carcinógenos/toxicidad , Herbicidas/toxicidad , Tumor de Células de Leydig/inducido químicamente , Testosterona/metabolismo , Animales , Expresión Génica/efectos de los fármacos , Humanos , Tumor de Células de Leydig/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Ratas , Medición de Riesgo , Testículo/efectos de los fármacos , Testículo/metabolismo , Testosterona/sangre
8.
Crit Rev Toxicol ; 44 Suppl 2: 1-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24832550

RESUMEN

Plant protection products (PPPs) and the active substance(s) contained within them are rigorously and comprehensively tested prior to registration to ensure that human health is not impacted by their use. In recent years, there has been a widespread drive to have more relevant testing strategies (e.g., ILSI/HESI-ACSA and new EU Directives), which also take account of animal welfare, including the 3R (replacement, refinement, and reduction) principles. The toxicity potential of one such new active substance, sulfoxaflor, a sulfoximine insecticide (CAS #946578-00-3), was evaluated utilizing innovative testing strategies comprising: (1) an integrated testing scheme to optimize information obtained from as few animals as possible (i.e., 3R principles) through modifications of standard protocols, such as enhanced palatability study design, to include molecular endpoints, additional neurotoxicity and immunotoxicity parameters in a subchronic toxicity study, and combining multiple test guidelines into one study protocol; (2) generation of toxicokinetic data across dose levels, sexes, study durations, species, strains and life stages, without using satellite animals, which was a first for PPP development, and (3) addition of prospective mode of action (MoA) endpoints within repeat dose toxicity studies as well as proactive inclusion of specific MoA studies as an integral part of the development program. These novel approaches to generate key data early in the safety evaluation program facilitated informed decision-making on the need for additional studies and contributed to a more relevant human health risk assessment. This supplement also contains papers which describe in more detail the approach taken to establish the MoA and human relevance framework related to toxicities elicited by sulfoxaflor in the mammalian toxicology studies: developmental toxicity in rats mediated via the fetal muscle nicotinic acetylcholine receptor (nAChR) ( Ellis-Hutchings et al. 2014 ); liver tumors in rodents mediated via CAR/PXR ( LeBaron et al. 2014 ); and Leydig cell tumors in Fischer 344 rats ( Rasoulpour et al. 2014 ).


Asunto(s)
Agroquímicos/toxicidad , Insecticidas/toxicidad , Piridinas/toxicidad , Compuestos de Azufre/toxicidad , Pruebas de Toxicidad/métodos , Bienestar del Animal , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Piridinas/farmacocinética , Medición de Riesgo , Compuestos de Azufre/farmacocinética
9.
Crit Rev Toxicol ; 44 Suppl 2: 15-24, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24832551

RESUMEN

Sulfoxaflor, a novel active substance that targets sap-feeding insects, induced rodent hepatotoxicity when administered at high dietary doses. Specifically, hepatocellular adenomas and carcinomas increased after 18 months in male and female CD-1 mice at 750 and 1250 ppm, respectively, and hepatocellular adenomas increased after 2 years in male F344 rats at 500 ppm. Studies to determine the mode of action (MoA) for these liver tumors were performed in an integrated and prospective manner as part of the standard battery of toxicology studies such that the MoA data were available prior to, or by the time of, the completion of the carcinogenicity studies. Sulfoxaflor is not genotoxic and the MoA data support the following key events in the etiology of the rodent liver tumors: (1) CAR nuclear receptor activation and (2) hepatocellular proliferation. The MoA data were evaluated in a weight of evidence approach using the Bradford Hill criteria for causation and were found to align with dose and temporal concordance, biological plausibility, coherence, strength, consistency, and specificity for a CAR-mediated MoA while excluding other alternate MoAs. The available data include: activation of CAR, Cyp2b induction, hepatocellular hypertrophy and hyperplasia, absence of liver effects in KO mice, absence of proliferation in humanized mice, and exclusion of other possible mechanisms (e.g., genotoxicity, cytotoxicity, AhR, or PPAR activation), and indicate that the identified rodent liver tumor MoA for sulfoxaflor would not occur in humans. In this case, sulfoxaflor is considered not to be a potential human liver carcinogen.


Asunto(s)
Insecticidas/toxicidad , Neoplasias Hepáticas/patología , Piridinas/toxicidad , Compuestos de Azufre/toxicidad , Animales , Carcinógenos/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Hepáticas/inducido químicamente , Masculino , Ratones , Ratas , Ratas Endogámicas F344 , Medición de Riesgo
10.
Crit Rev Toxicol ; 44 Suppl 2: 25-44, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24832552

RESUMEN

Sulfoxaflor, a molecule that targets sap-feeding insects, was assessed for carcinogenic potential in groups of 50 Fischer rats fed with diets containing 0, 25, 100, 500 (males), or 750 (females) ppm sulfoxaflor for 2 years according to OECD 453. Sulfoxaflor did not alter the number of rats with Leydig cell tumors (LCTs: 88% of controls and 90-92% in treated groups). The size of LCT was increased at 100 and 500 ppm. The spontaneous incidence of LCT in Fischer rat is 75-100% compared with less than 0.01% in humans. These fundamental interspecies differences in spontaneous incidence of LCT are the result of quantitative and qualitative differences in Leydig cell response to hormonal stimuli. There are nine known modes of actions (MoA) for LCT induction. Analysis sulfoxaflor data suggested a hormone-based dopamine enhancement MoA causing the LCT effect through: 1) increased neuronal dopamine release via specific dopaminergic neuron-based nicotinic acetylcholine receptor (nAChR) agonism, leading to 2) decreased serum prolactin (Prl) levels, 3) downregulation of luteinizing hormone receptor (LHR) gene expression in Leydig cells, 4) transient decreases in serum testosterone, 5) increased serum LH levels, and 6) promotion of LCTs. The analysis suggested that sulfoxaflor promoted LCTs through a subtle stimulation of dopamine release. The MoA for LCT promotion in the carcinogenicity study is considered to have no relevance to humans due to qualitative and quantitative differences between rat and human Leydig cells. Therefore, the Fischer 344 rat LCT promotion associated with lifetime administration of high-dose levels of sulfoxaflor would not pose a cancer hazard to humans.


Asunto(s)
Tumor de Células de Leydig/patología , Piridinas/toxicidad , Compuestos de Azufre/toxicidad , Animales , Carcinógenos/toxicidad , Modelos Animales de Enfermedad , Humanos , Tumor de Células de Leydig/inducido químicamente , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/patología , Masculino , Ratas
11.
Crit Rev Toxicol ; 44 Suppl 2: 45-62, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24832553

RESUMEN

Sulfoxaflor (CAS# 946578-00-3) is a novel active substance with insecticidal properties mediated via its agonism on the highly abundant insect nicotinic acetylcholine receptor (nAChR). In developmental and reproductive toxicity studies, gestational exposure caused fetal abnormalities (primarily limb contractures) and reduced neonatal survival in rats, but not rabbits, following high-dose dietary exposure. Sulfoxaflor induced these effects via a novel mode of action (MoA) mediated by the fetal-type muscle nAChR with the following key events: (1) binding to the receptor, (2) agonism on the receptor, causing (3) sustained muscle contracture in the near-term fetus and neonatal offspring. This sustained muscle contracture results in misshapen limbs, bent clavicles, and reduced diaphragm function, which compromises respiration in neonatal rats at birth, reducing their survival. This review evaluates the weight of evidence for this MoA based upon the Bradford Hill criteria, includes a cross-comparison of applied and internal doses in a variety of in vitro, ex vivo, and in vivo study designs, examines alternative MoAs, and applies a Human relevance framework (HRF) to ascertain human risk for this rat MoA. The review indicated, with a high level of confidence, that the sulfoxaflor-induced fetal abnormalities and neonatal death in rats occur via a single MoA comprising sustained activation of the rat fetal-type muscle nAChR resulting in a sustained muscle contracture. This MoA is considered not relevant to humans, given fundamental qualitative differences in sulfoxaflor agonism on the rat versus the human muscle nAChR. Specifically, sulfoxaflor does not cause agonism on either the human fetal- or adult-type muscle nAChR.


Asunto(s)
Insecticidas/toxicidad , Piridinas/toxicidad , Compuestos de Azufre/toxicidad , Animales , Carcinógenos/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Contracción Muscular/efectos de los fármacos , Proteínas Musculares/metabolismo , Ratas , Receptores Nicotínicos/metabolismo , Reproducción/efectos de los fármacos
12.
Toxicol Appl Pharmacol ; 270(2): 164-73, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23607986

RESUMEN

Registration of new plant protection products (e.g., herbicide, insecticide, or fungicide) requires comprehensive mammalian toxicity evaluation including carcinogenicity studies in two species. The outcome of the carcinogenicity testing has a significant bearing on the overall human health risk assessment of the substance and, consequently, approved uses for different crops across geographies. In order to understand the relevance of a specific tumor finding to human health, a systematic, transparent, and hypothesis-driven mode of action (MoA) investigation is, appropriately, an expectation by the regulatory agencies. Here, we describe a novel approach of prospectively generating the MoA data by implementing additional end points to the standard guideline toxicity studies with sulfoxaflor, a molecule in development. This proactive MoA approach results in a more robust integration of molecular with apical end points while minimizing animal use. Sulfoxaflor, a molecule targeting sap-feeding insects, induced liver effects (increased liver weight due to hepatocellular hypertrophy) in an initial palatability probe study for selecting doses for subsequent repeat-dose dietary studies. This finding triggered the inclusion of dose-response investigations of the potential key events for rodent liver carcinogenesis, concurrent with the hazard assessment studies. As predicted, sulfoxaflor induced liver tumors in rats and mice in the bioassays. The MoA data available by the time of the carcinogenicity finding supported the conclusion that the carcinogenic potential of sulfoxaflor was due to CAR/PXR nuclear receptor activation with subsequent hepatocellular proliferation. This MoA was not considered to be relevant to humans as sulfoxaflor is unlikely to induce hepatocellular proliferation in humans and therefore would not be a human liver carcinogen.


Asunto(s)
Insecticidas/toxicidad , Hígado/efectos de los fármacos , Piridinas/toxicidad , Compuestos de Azufre/toxicidad , Pruebas de Toxicidad/métodos , Animales , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP2B1/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Insecticidas/administración & dosificación , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Estudios Prospectivos , Piridinas/administración & dosificación , ARN/química , ARN/genética , Ratas , Ratas Endogámicas F344 , Reacción en Cadena en Tiempo Real de la Polimerasa , Compuestos de Azufre/administración & dosificación
13.
Birth Defects Res B Dev Reprod Toxicol ; 95(2): 107-15, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22461433

RESUMEN

N-(2-aminoethyl)ethanolamine (AEEA) caused aneurysms of the great vessels in rats exposed in utero and during the first days post partum, exacerbated by postnatal treatment of the lactating dams (Moore et al., 2012). The purpose of this work was to examine the systemic availability of AEEA during gestation and early lactation. The absorption of AEEA was determined following oral administration to nonpregnant and pregnant female Wistar rats. A single dose administered by gavage (0.5 or 50 mg/kg) on gestation day 18 was rapidly and extensively (>90%) absorbed from the gastrointestinal tract (absorption t(1/2) = 0.1-0.2 hr). Elimination from the plasma followed a biphasic pattern, with a rapid elimination phase (t(1/2 α) = 1.6-1.8 hr) followed by a slower phase (t(1/2 ß) = 16.7-17.3 hr). Following repeated gavage administration during gestation day 17 to 19, (14) C-AEEA-derived radioactivity readily partitioned into the fetus and was evenly distributed therein, but cleared approximately twofold slower from the fetal blood and tissues than the maternal blood and chorioallantoic placenta. When administered to lactating dams during lactation days 1 to 12, (14) C-AEEA-derived radioactivity preferentially partitioned into the milk reaching levels that were between 1.6- and 2.5-fold higher than the maternal blood. Although the concentration of AEEA equivalents in the maternal blood remained quite consistent, the concentration in the milk fell by almost 40% between lactation days 4 and 12, probably reflecting an increase in milk production over this same period. We confirm exposure of the offspring to AEEA both in utero and during lactation, but that AEEA does not appear to specifically concentrate in the great vessels.


Asunto(s)
Etanolaminas/farmacocinética , Etanolaminas/toxicidad , Feto/efectos de los fármacos , Leche/química , Efectos Tardíos de la Exposición Prenatal , Administración Oral , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Femenino , Lactancia , Exposición Materna , Intercambio Materno-Fetal , Embarazo , Ratas , Ratas Wistar , Distribución Tisular
14.
Regul Toxicol Pharmacol ; 62(3): 474-81, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22178771

RESUMEN

Diiodomethyl-p-tolylsulfone (DIMPTS) was tested in developmental toxicity (DT) and reproductive toxicity studies. In the rat DT study, DIMPTS was administered at 0, 100, 300 or 1000 mg/kg/day. Maternal toxicity as evidenced by reductions in body weight gain or feed consumption at 1000 and, to a lesser extent, 300 mg/kg/day. The only developmental effect was umbilical hernia at 1000 mg/kg/day; therefore, NOELs for maternal and developmental toxicity were 100 and 300 mg/kg/day, respectively. In the rabbit DT study, NZW rabbits were gavaged with 0, 0.05, 0.5 and 2mg/kg/day DIMPTS. The NOEL for maternal toxicity was 0.5mg/kg/day, based on thyroid weight increase with histopathology. There were no observed developmental effects. In the two-generation study, CD rats were fed 0, 2.5, 10 or 40 mg/kg/day DIMPTS. Increased thyroid weight and histopathology were observed at all doses with associated pituitary findings in males. Reproductive toxicity at 40 mg/kg/day consisted of increased postimplantation loss, decreased gestation survival and two cases of dystocia, while litter size, pup survival/weight were affected at 10 and 40 mg/kg/day. The NOEL for parental toxicity could not be determined, while the NOEL for reproductive toxicity was 2.5mg/kg/day. The maternal thyroid and reproductive effects in this study were consistent with iodine toxicity.


Asunto(s)
Anomalías Inducidas por Medicamentos/diagnóstico , Derivados del Benceno/toxicidad , Desarrollo Fetal/efectos de los fármacos , Reproducción/efectos de los fármacos , Sulfonas/toxicidad , Anomalías Inducidas por Medicamentos/patología , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Femenino , Desarrollo Fetal/fisiología , Masculino , Embarazo , Conejos , Ratas , Reproducción/fisiología , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/crecimiento & desarrollo , Glándula Tiroides/patología
15.
Regul Toxicol Pharmacol ; 63(2): 321-32, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22440553

RESUMEN

Integrated toxicokinetics (TK) data provide information on the rate, extent and duration of systemic exposure across doses, species, strains, gender, and life stages within a toxicology program. While routine for pharmaceuticals, TK assessments of non-pharmaceuticals are still relatively rare, and have never before been included in a full range of guideline studies for a new agrochemical. In order to better understand the relationship between diurnal systemic dose (AUC(24h)) and toxicity of agrochemicals, TK analyses in the study animals is now included in all short- (excluding acute), medium- and long-term guideline mammalian toxicity studies including reproduction/developmental tests. This paper describes a detailed procedure for the implementation of TK in short-, medium- and long-term regulatory toxicity studies, without the use of satellite animals, conducted on three agrochemicals (X11422208, 2,4-D and X574175). In these studies, kinetically-derived maximum doses (KMD) from short-term studies instead of, or along with, maximum tolerated doses (MTD) were used for the selection of the high dose in subsequent longer-term studies. In addition to leveraging TK data to guide dose level selection, the integrated program was also used to select the most appropriate method of oral administration (i.e., gavage versus dietary) of test materials for rat and rabbit developmental toxicity studies. The integrated TK data obtained across toxicity studies (without the use of additional/satellite animals) provided data critical to understanding differences in response across doses, species, strains, sexes, and life stages. Such data should also be useful in mode of action studies and to improve human risk assessments.


Asunto(s)
Agroquímicos , Alternativas al Uso de Animales/métodos , Pruebas de Toxicidad/métodos , Agroquímicos/administración & dosificación , Agroquímicos/farmacocinética , Agroquímicos/toxicidad , Alternativas al Uso de Animales/estadística & datos numéricos , Animales , Simulación por Computador , Interpretación Estadística de Datos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Guías como Asunto , Humanos , Dosis Máxima Tolerada , Valor Predictivo de las Pruebas , Conejos , Ratas , Ratas Endogámicas F344 , Reproducción/efectos de los fármacos , Pruebas de Toxicidad/estadística & datos numéricos
16.
Toxicol Mech Methods ; 21(4): 298-311, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21495868

RESUMEN

There has been a growing concern that epigenetic events, that is, heritable changes in gene expression superimposed on DNA nucleotide sequences, may be involved in chemically and/or nutritionally mediated adverse health outcomes, such as reproductive toxicity and cancer. This concern has been driven by an increasing number of studies reporting toxicant-induced alterations to the epigenome in the form of changes in DNA methylation, histone/chromatin remodeling, and altered expression of non-coding RNAs. These three major mechanisms of epigenetic modifications may have coordinated, independent, or potentially antagonistic influences on gene expression. Complicating this understanding is the incomplete understanding of the normal state and dynamic variation of the epigenome, which differs widely between cells, tissues, developmental state, age, strain, and species. This review serves as a framework to outline characteristics composing an ideal epigenetic screen(s) for hazard identification in product safety assessment. In order to implement such a screen, first there needs to be a better understanding of adaptive versus adverse changes in the epigenome, which includes identification of robust and reproducible causal links between epigenetic changes and adverse apical end points, and second development of improved reporter assay tools to monitor such changes. An ideal screen would be in vitro-based, medium- to high-throughput, and assess all three branches of epigenome control (i.e. methylation, histone modifications, non-coding RNAs), although also being quantitative, objective, portable (i.e. lab to lab), and relevant to humans.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inducido químicamente , Epigénesis Genética/efectos de los fármacos , Toxicología/métodos , Alternativas a las Pruebas en Animales , Animales , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Disruptores Endocrinos/toxicidad , Marcadores Genéticos , Impresión Genómica/efectos de los fármacos , Impresión Genómica/genética , Histonas/genética , Humanos , Medición de Riesgo , Pruebas de Toxicidad
17.
Front Toxicol ; 3: 766196, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35295143

RESUMEN

Nitrapyrin, a nitrification inhibitor, produces liver tumors in B6C3F1 mice. In a 2-year oncogenicity study, increased incidence of mice with hepatocellular tumors was observed following exposure to 125 (females only) or 250 mg/kg/day (males and females) nitrapyrin in the diet. Previous data was generated in male mice to support a mode-of-action (MoA) characterized by constitutive androstane receptor (CAR) nuclear receptor (NR) activation, increased hepatocellular proliferation, and subsequent hepatocellular foci and tumor formation. Uncertainty as to the relevance of this MoA for females remained given the increased sensitivity to tumor formation in female mice. A targeted MoA study was conducted to evaluate CAR activation and hepatic responses in female mice treated with the female carcinogenic dose of nitrapyrin for 4 days. Nitrapyrin induced a treatment-related increase in hepatocellular hypertrophy and hepatocellular proliferation. Nitrapyrin also induced a dose-related increase in the Cyp2b10/CAR-associated transcript and liver weights. Nitrapyrin-induced liver weights and Cyp2b10 gene expression for both males and females were compared to data generated from three other established CAR activators; methyl isobutyl ketone, phenobarbital, and sulfoxaflor. The response observed in female mice following exposure to nitrapyrin was within range of the degree of change observed in mice following exposure to tumorigenic doses of other CAR activators. Consistent with the liver MoA in male mice, these data support a CAR-mediated mode of action for nitrapyrin-induced liver tumors in female mice, with the understanding that a focused approach minimizing animal use can bridge male and female datasets when sex-specific carcinogenic differences are observed.

18.
Mutat Res ; 705(2): 83-95, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20399890

RESUMEN

Epigenetics, as it pertains to biology and toxicology, can be defined as heritable changes in gene expression that do not involve mutations and are propagated without continued stimulus. Although potentially reversible, these heritable changes may be classified as mitotic, meiotic, or transgenerational, implicating the wide-ranging impact of epigenetic control in cellular function. A number of biological responses have been classified as being caused by an "epigenetic alteration," sometimes based on sound scientific evidence and often in lieu of an identified genetic mutation. Complicating the understanding and interpretation of perceived epigenetic alterations is an incomplete understanding of the normal state and dynamic variation of the epigenome, which can differ widely between cell and tissue types and stage of development or age. This emerging field is likely to have a profound impact on the study and practice of toxicology in coming years. This document reviews the current state of the science in epigenetic modifications, techniques used to measure these changes, and evaluates the current toxicology testing battery with respect to strengths and potential weaknesses in the identification of epigenetics changes. In addition, case studies implicating transgenerational effects induced by diethylstilbestrol, vinclozolin, and bisphenol A were reviewed to illustrate the application of epigenetics in safety assessment and the strengths and limitations of the study designs. An assessment of toxicology tests currently used in safety evaluation revealed that these tests are expected to identify any potential adverse outcomes resulting from epigenetic changes. Furthermore, in order to increase our understanding of the science of epigenetics in toxicology, this review has revealed that a solid understanding of the biology and variation in the epigenome is essential to contextualize concerns about possible adverse health effects related to epigenetic changes. Finally, the fundamental principles guiding toxicology studies, including relevant doses, dose-rates, routes of exposure, and experimental models, need to be taken into consideration in the design and interpretation of studies within this emerging area of science.


Asunto(s)
Epigénesis Genética , Pruebas de Toxicidad , Animales , Metilación de ADN , Perros , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Inmunidad/efectos de los fármacos , Ratones , Modelos Animales , Reproducción/efectos de los fármacos , Medición de Riesgo , Toxicología
19.
Birth Defects Res B Dev Reprod Toxicol ; 83(6): 573-81, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19025792

RESUMEN

High-dose gavage exposure to ethylene glycol (EG) is teratogenic in rats, but not rabbits. To investigate the reason for this species difference, toxicokinetic and whole embryo culture (WEC) studies were conducted in gestation day 9 New Zealand White rabbits, and the data compared to very similar data previously generated in pregnant rats. In the toxicokinetic study, maximal levels of unchanged EG in rabbits were comparable to those reported for rats. However, maximal levels of EG's teratogenic metabolite, glycolic acid (GA), in rabbit maternal blood and embryo were only 46% and 10% of the respective levels in rats. The toxicokinetic profile suggested that the lower GA levels in rabbits were due to a slower rate of maternal metabolism of EG to GA, slow uptake of GA into the yolk sac cavity fluid which surrounds the embryo, and negligible transfer via the visceral yolk sac (VYS) placenta. In the WEC study, exposure of rabbit conceptuses to high concentrations (< or = 12.5 mM) of GA was without effect, which contrasts with reported effects in rat WEC at > or = 3 mM. Overall, these data implicate toxicokinetics as an important factor underlying the species difference, although intrinsic insensitivity of the rabbit embryo might also be involved. Integration of these findings with published human data suggest that the rabbit is the more relevant model for human EG exposure, based on the negligible role of the rabbit VYS in placental transfer (humans lack a VYS) and similar rates of EG metabolism and extraembryonic fluid turnover.


Asunto(s)
Anomalías Inducidas por Medicamentos/patología , Embrión de Mamíferos/efectos de los fármacos , Glicol de Etileno/toxicidad , Feto/efectos de los fármacos , Técnicas de Cultivo de Órganos , Teratógenos/toxicidad , Animales , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Glicol de Etileno/farmacocinética , Femenino , Feto/anomalías , Humanos , Masculino , Embarazo , Conejos , Especificidad de la Especie , Teratógenos/farmacocinética
20.
Biol Reprod ; 76(2): 279-85, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17123945

RESUMEN

The transcription factor NF-kappaB modulates apoptotic machinery following activation by the IkappaB kinase (IKK) complex. Inhibiting activity of one of the catalytic subunits of the IKK complex, IKKbeta (also known as IKBKB and IKK2) severely inhibits NF-kappaB nuclear translocation in response to most stimuli, including ionizing radiation. Doubly floxed Ikkbeta(F/F) mice (control) were compared to haplo-insufficient Ikkbeta(F/)(delta) mice (NF-kappaB knockdown) to examine the in vivo apoptotic role of NF-kappaB in the testis. Although Ikkbeta(F/F) control adult mice had spermatid head counts and testis and body weights similar to Ikkbeta(F/)(delta) mice, cellular stress in the form of ionizing radiation elicited a differential phenotype. Lower body exposure to 5 Gy of ionizing radiation induced a greater NF-kappaB activation in Ikkbeta(F/F) than in Ikkbeta(F/)(delta) mice. In addition, exposure to ionizing radiation resulted in fewer apoptotic germ cells 3, 6, and 12 h after injury in NF-kappaB knockdown mice than in controls, concomitant with the reduced cleavage of caspases 3 and 9 at 3 h. There was also a reduction in total germ cells lost after radiation with NF-kappaB inhibition. Correspondingly, real-time RT-PCR showed a significant reduction in Cdnk1a (also known as p21) and Fasl expression 3 and 6 h, respectively, after irradiation in Ikkbeta(F/)(delta) compared to control testes. These data indicate that, despite acting in an antiapoptotic manner in many tissue types, NF-kappaB is proapoptotic in modulating the germ cell response to ionizing radiation.


Asunto(s)
Apoptosis/fisiología , Rayos gamma , FN-kappa B/metabolismo , FN-kappa B/efectos de la radiación , Espermatozoides/fisiología , Testículo/fisiología , Animales , Caspasas/metabolismo , Susceptibilidad a Enfermedades , Activación Enzimática , Perfilación de la Expresión Génica , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Masculino , Ratones , Ratones Noqueados , FN-kappa B/deficiencia , Fenotipo , ARN/metabolismo , Traumatismos por Radiación/metabolismo , Testículo/citología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA