Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Food Chem Toxicol ; 150: 112084, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33621607

RESUMEN

Bis-hexanoyl (R)-1,3-butanediol (BH-BD) is novel ketone ester undergoing development as a food ingredient to achieve nutritional ketosis in humans. Male and female Crl:CD(SD) rats were administered BH-BD twice daily at 9000, 12,000 or 15,000 mg/kg/day, by oral gavage in a 90-day toxicity study with 28-day recovery period; and an interim 28-day phase. Test substance-related early deaths occurred in four females at 15,000 mg/kg/day. A dose-dependent increase in acute transient postdose (1-3 h) observations of incoordination at ≥12,000 mg/kg/day and decreased activity at all dose levels were noted in both sexes. Postdose observations were likely associated with peak ketonemia and were considered adverse at 15,000 mg/kg/day. These daily observations decreased over the study without any persistent effects, as determined during weekly pre-dose observations. Adverse histopathological changes included ulceration/erosion in non-glandular stomach at ≥ 12,000 mg/k/day and in glandular stomach at 15,000 mg/kg/day. These histopathological findings were not noted after 28-days of recovery. Due to unlikely human relevance of the rat non-glandular stomach effects for BH-BD and test substance-related mortality at 15,000 mg/kg/day, the no-observed-adverse-effect level (NOAEL) for subchronic toxicity of BH-BD was determined to be 12,000 mg/kg/day.


Asunto(s)
Butileno Glicoles/toxicidad , Animales , Butileno Glicoles/química , Esquema de Medicación , Femenino , Masculino , Estructura Molecular , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Pruebas de Toxicidad Subcrónica
4.
Birth Defects Res ; 111(1): 26-33, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30511437

RESUMEN

BACKGROUND: To protect crewmember health, the U.S. Navy sets exposure limits for more than 200 components of submarine atmospheres. The addition of females to nuclear submarines required a reevaluation of these exposure limits, originally established for all-male crews. In the case of carbon dioxide (CO2 ), the only available data suitable for deriving an exposure limit were from a 2010 study sponsored by the British Royal Navy that reported a debatable interpretation casting doubt on whether current U.S. Navy exposure limits served to protect fetal developmental health. METHODS: About 120 time-mated female Sprague-Dawley rats (Crl: CD[SD]) were exposed to CO2 at levels of 1.5%, 2.0%, 2.5%, and 3.0% from gestation days 6 to 20. Dams were euthanized and fetuses were examined. RESULTS: Findings with implications for exposure limits for CO2 during pregnancy were an increased mean litter proportion of early resorptions and a lower mean litter proportion of viable fetuses in the 3.0% CO2 group. CONCLUSION: The results yield a No Observed Adverse Effect Level (NOAEL) of 2.5% and a Lowest Observed Adverse Effect Level (LOAEL) of 3.0%. The results reasonably allow a point of departure of 2.5% CO2 for deriving an exposure recommendation. An interspecies uncertainty factor was applied to derive a recommended 90-day continuous exposure limit (CEL) of 0.8% for CO2 . As reproductive endpoints that are developmental in nature must be assumed to result from a single exposure at a critical point during gestation, it is further recommended that the 24-hr emergency exposure limit (EEL) also be 0.8%.


Asunto(s)
Dióxido de Carbono/toxicidad , Medicina Submarina/normas , Animales , Peso Corporal/efectos de los fármacos , Dióxido de Carbono/metabolismo , Modelos Animales de Enfermedad , Femenino , Desarrollo Fetal/efectos de los fármacos , Feto/efectos de los fármacos , Personal Militar , Nivel sin Efectos Adversos Observados , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos , Medicina Submarina/métodos
5.
Toxicol Pathol ; 43(3): 343-53, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25107574

RESUMEN

Histopathologic examination of the immature ovary is a required end point on juvenile toxicity studies and female pubertal and thyroid function assays. To aid in this evaluation and interpretation of the immature ovary, the characteristic histologic features of rat ovary through the developmental periods are described. These histologic features are correlated with published changes in neuroendocrine profiles as the hypothalamic-pituitary-gonadal axis matures. During the neonatal stage (postnatal day [PND] 0-7), ovarian follicle development is independent of pituitary gonadotropins (luteinizing hormone [LH] or follicle-stimulating hormone [FSH]), and follicles remain preantral. Antral development of "atypical" follicles occurs in the early infantile period (PND 8-14) when the ovary becomes responsive to pituitary gonadotropins. In the late infantile period (PND 15-20), the zona pellucida appears, the hilus forms, and antral follicles mature by losing their "atypical" appearance. The juvenile stage (PND 21-32) is the stage when atresia of medullary follicles occurs corresponding to a nadir in FSH levels. In the peripubertal period (PND 33-37), atresia subsides as FSH levels rebound, and LH begins its bimodal surge pattern leading to ovulation. This report will provide pathologists with baseline morphologic and endocrinologic information to aid in identification and interpretation of xenobiotic effects in the ovary of the prepubertal rat.


Asunto(s)
Ovario/anatomía & histología , Ovario/crecimiento & desarrollo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Estro/fisiología , Femenino , Hormonas Esteroides Gonadales/sangre , Sistemas Neurosecretores/crecimiento & desarrollo , Sistemas Neurosecretores/fisiología , Folículo Ovárico/fisiología , Ovario/fisiología , Ratas , Ratas Sprague-Dawley
6.
Toxicol Pathol ; 43(3): 326-42, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25217330

RESUMEN

Histopathologic examination of the testis from juvenile rats is often necessary to characterize the safety of new drugs for pediatric use and is a required end point in male pubertal development and thyroid function assays. To aid in evaluation and interpretation of the immature testis, the characteristic histologic features of the developing rat testis throughout postnatal development are described and correlated with published neuroendocrine parameter changes. During the neonatal period (postnatal day [PND] 3-7), seminiferous tubules contained gonocytes and mitotically active immature Sertoli cells. Profound proliferation of spermatogonia and continued Sertoli cell proliferation occurred in the early infantile period (PND 8-14). The spermatogonia reached maximum density forming double-layered rosettes with Sertoli cells in the late infantile period (PND 15-20). Leptotene/zygotene spermatocytes appeared centrally as tubular lumina developed, and individual tubules segregated into stages. The juvenile period (PND 21-32) featured a dramatic increase in number and size of pachytene spermatocytes with the formation of round spermatids and loss of "infantile" rosette architecture. In the peri-pubertal period (PND 32-55), stage VII tubules containing step 19 spermatids were visible by PND 46. The presented baseline morphologic and endocrinologic information will help pathologists distinguish delayed development from xenobiotic effects, determine pathogenesis when confronted with nonspecific findings, and identify sensitive time points for targeted study design.


Asunto(s)
Sistemas Neurosecretores/crecimiento & desarrollo , Sistemas Neurosecretores/fisiología , Testículo/anatomía & histología , Testículo/crecimiento & desarrollo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Peso Corporal/fisiología , Hormonas Esteroides Gonadales/sangre , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Inmunohistoquímica , Masculino , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Espermatogonias/patología , Testículo/fisiología
7.
Birth Defects Res B Dev Reprod Toxicol ; 101(1): 43-62, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24510766

RESUMEN

The male and female pubertal assays, which are included in the U.S. Environmental Protection Agency's (EPA) Endocrine Disruptor Screening Program (EDSP) Tier 1 battery, can detect endocrine-active compounds operating by various modes of action. This article uses the collective experience of three laboratories to provide information on pubertal assay conduct, interlaboratory reproducibility, endpoint redundancy, and data interpretation. The various criteria used to select the maximum tolerated dose are described. A comparison of historical control data across laboratories confirmed reasonably good interlaboratory reproducibility. With a reliance on apical endpoints, interpretation of pubertal assay effects as specifically endocrine-mediated or secondary to other systemic effects can be problematic and mode of action may be difficult to discern. Across 21-23 data sets, relative liver weight, a nonspecific endocrine endpoint, was the most commonly affected endpoint in male and female assays. For endocrine endpoints, patterns of effects were generally seen; rarely was an endocrine-sensitive endpoint affected in isolation. In males, most frequently missed EPA-established performance criteria included mean weights for kidney and thyroid, and the coefficient of variation for age and body weight at preputial separation, seminal vesicle weight, and final body weight. In females, the frequently missed EPA-established performance criteria included mean adrenal weight and mean age at vaginal opening. To ensure specificity for endocrine effects, the pubertal assays should be interpreted using a weight-of-evidence approach as part of the entire EDSP battery. Based on the frequency with which certain performance criteria were missed, an EPA review of these criteria is warranted.


Asunto(s)
Bioensayo/métodos , Disruptores Endocrinos/análisis , Disruptores Endocrinos/toxicidad , Maduración Sexual/efectos de los fármacos , Pruebas de Toxicidad/métodos , United States Environmental Protection Agency , Animales , Determinación de Punto Final , Ciclo Estral , Femenino , Masculino , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Estados Unidos
8.
J Immunotoxicol ; 11(4): 376-82, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24219298

RESUMEN

Recently, there has been a renewed interest in the use of the minipig as an alternative to dogs and non-human primates for conducting toxicological assessments in non-rodent species. Since the T-dependent antibody response (TDAR) is one of the most widely-accepted assays used in the assessment of immunocompetence, the present study was undertaken to characterize the primary and secondary TDAR to keyhole limpet hemocyanin (KLH) in the Göttingen Minipig(®). Following primary immunization with either 2 or 10 mg KLH, anti-swine IgM and IgG ELISAs were optimized and individual animal responses were evaluated over time. Immunization with 10 mg KLH on Day 0 promoted primary IgM responses that peaked 6-9 days after antigen administration, while primary IgG levels peaked on Day 13 or 14. Secondary IgG antibody levels (following secondary injection with 2 mg KLH on Day 14) plateaued on Days 20-22. Anti-KLH antibody levels were decreased in minipigs treated with cyclophosphamide (CPS), a known immunosuppressant, at doses ranging from 12.5-50 mg/kg/day, while antibody levels in animals treated with 2.5 mg CPS/kg/day were similar to levels in saline-treated swine. These results demonstrate that the Göttingen Minipig(®) can be a useful alternative non-rodent species to the dog and the non-human primate for evaluating the TDAR to KLH in regulatory assessments of immunotoxicity.


Asunto(s)
Modelos Animales , Porcinos Enanos , Animales , Formación de Anticuerpos/efectos de los fármacos , Ciclofosfamida/administración & dosificación , Perros , Hemocianinas/inmunología , Inmunización Secundaria , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Inmunosupresores/administración & dosificación , Primates , Porcinos , Linfocitos T Colaboradores-Inductores/inmunología
9.
Artículo en Inglés | MEDLINE | ID: mdl-21284075

RESUMEN

BACKGROUND: Decabromodiphenyl ether (DecaBDE; CASRN 1163-19-5) is a flame retardant used in a variety of manufactured products. A single oral dose of 20.1 mg/kg administered to mice on postnatal day 3 has been reported to alter motor activity at 2, 4, and 6 months of age. METHODS: To further evaluate these results, a developmental neurotoxicity study was conducted in the most commonly used species for studies of this type, the rat, according to international validated testing guidelines and Good Laboratory Practice Standards. DecaBDE was administered orally via gavage in corn oil to dams from gestation day 6 to weaning at doses of 0, 1, 10, 100, or 1,000 mg/kg/day. Standard measures of growth, development, and neurological endpoints were evaluated in the offspring. Motor activity was assessed at 2 months of age. Additional motor activity assessments were conducted at 4 and 6 months of age. Neuropathology and morphometry evaluations of the offspring were performed at weaning and adulthood. RESULTS: No treatment-related neurobehavioral changes were observed in detailed clinical observations, startle response, or learning and memory tests. No test substance-related changes were noted in motor activity assessments performed at 2, 4, or 6 months of age. Finally, no treatment-related neuropathological or morphometric alterations were found. CONCLUSIONS: Under the conditions of this study, the no-observed-adverse-effect level for developmental neurotoxicity of DecaBDE was 1,000 mg/kg/day, the highest dose tested.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Éteres Difenilos Halogenados/administración & dosificación , Éteres Difenilos Halogenados/toxicidad , Neurotoxinas/toxicidad , Efectos Tardíos de la Exposición Prenatal/patología , Pruebas de Toxicidad , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Cruzamientos Genéticos , Conducta Alimentaria/efectos de los fármacos , Femenino , Masculino , Memoria/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Neurotoxinas/administración & dosificación , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Análisis de Supervivencia , Natación
10.
Drug Metab Dispos ; 38(10): 1648-54, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20581093

RESUMEN

The effects of route and vehicle on blood and milk levels of decabromodiphenyl ether (DecaBDE; CASRN 1163-19-5) were investigated in the rat to assist in the design and conduct of a developmental neurotoxicity study. Blood plasma and/or milk concentrations were determined in dams, fetuses, and/or nursing pups after repeated DecaBDE administration by gavage throughout gestation or gestation and lactation using corn oil (CO) or soyaphospholipon/Lutrol F 127-water (SPL) as the vehicle. The impact of vehicle on plasma levels was also investigated in pups derived from naive dams after a single postnatal dose. This study reports for the first time fetal and neonatal plasma concentrations concurrent with those of maternal plasma and/or milk. Higher concentrations of DecaBDE were achieved in plasma and in milk with CO than with SPL. Furthermore, pups derived from dams treated with only SPL were lower in body weight, compared with those from dams treated with either CO, CO and DecaBDE, or SPL and DecaBDE. The study further shows that exposure to DecaBDE is relatively consistent across the dose range of 100 to 1000 mg/(kg · day) when administered in CO.


Asunto(s)
Sangre Fetal/metabolismo , Retardadores de Llama/farmacocinética , Éteres Difenilos Halogenados/sangre , Exposición Materna/efectos adversos , Leche/metabolismo , Pruebas de Toxicidad/métodos , Administración Oral , Animales , Animales Recién Nacidos , Aceite de Maíz/química , Relación Dosis-Respuesta a Droga , Femenino , Retardadores de Llama/toxicidad , Edad Gestacional , Éteres Difenilos Halogenados/farmacocinética , Éteres Difenilos Halogenados/toxicidad , Intercambio Materno-Fetal , Polietilenos/química , Polipropilenos/química , Embarazo , Ratas , Ratas Sprague-Dawley
11.
Reprod Toxicol ; 29(3): 271-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20184952

RESUMEN

Spontaneous ejaculation has been reported in a variety of mammalian species and may occur either as a result of pharmacological treatment or as a component of the daily behavior of normal, untreated animals. Infrequently, increased numbers of spontaneous ejaculatory plugs have been reported among the clinical signs in rat toxicology studies. This mini-review presents an overview on the presence of ejaculatory plugs in rodents and provides recommendations to consider when attempting to understand the toxicological significance of increased numbers of ejaculatory plugs in rat toxicology studies.


Asunto(s)
Eyaculación/fisiología , Fenómenos Fisiológicos , Animales , Masculino , Observación , Ratas
12.
Neurotoxicology ; 31(3): 247-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20171981

RESUMEN

The potential for neurotoxicological and immunotoxicological effects of ethylbenzene was studied in young adult Crl:CD(SD) rats following 90-day oral (neurotoxicity) or 28-day inhalation (immunotoxicity) exposures. In the neurotoxicity study, ethylbenzene was administered orally via gavage twice daily at 0, 25, 125, or 250 mg/kg per dose (total daily dosages of 0, 50, 250, or 500 mg/kg bwt/day [mg/kg bwt/day]) for 13 weeks and the functional observational battery (FOB), automated tests for motor activity and neuropathological examination were conducted. In the immunotoxicity study, animals were exposed by inhalation to 0, 25, 100, or 500 ppm ethylbenzene (approximately 26, 90, or 342 mg/kg bwt/day as calculated from physiologically based pharmacokinetic modeling). Immunotoxicity was evaluated in female rats using the splenic antibody-forming cell plaque-forming assay in sheep red blood cell sensitized animals. The no-observed-effect level for the oral gavage study was 50mg/kg bwt/day based on increased relative weights of the liver and kidneys in the male rats. The no-observed-adverse-effect level (NOAEL) for adult neurotoxicity was the highest dose tested 500 mg/kg bwt/day. The NOAEL for the immunotoxicity evaluation was the highest tested exposure concentration, 500 ppm (342 mg/kg bwt/day).


Asunto(s)
Derivados del Benceno/toxicidad , Modelos Animales de Enfermedad , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/inmunología , Neurotoxinas/toxicidad , Administración por Inhalación , Administración Oral , Análisis de Varianza , Animales , Derivados del Benceno/administración & dosificación , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Enfermedades Renales/inducido químicamente , Hepatopatías/etiología , Masculino , Actividad Motora/fisiología , Examen Neurológico/métodos , Síndromes de Neurotoxicidad/complicaciones , Síndromes de Neurotoxicidad/mortalidad , Nivel sin Efectos Adversos Observados , Oftalmología , Ratas , Ratas Sprague-Dawley , Factores Sexuales
13.
Toxicol Sci ; 115(1): 167-82, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20164145

RESUMEN

This study was conducted to determine the potential of bisphenol A (BPA) to induce functional and/or morphological effects to the nervous system of F(1) offspring from dietary exposure during gestation and lactation according to the Organization for Economic Cooperation and Development and U.S. Environmental Protection Agency guidelines for the study of developmental neurotoxicity. BPA was offered to female Sprague-Dawley Crl:CD (SD) rats (24 per dose group) and their litters at dietary concentrations of 0 (control), 0.15, 1.5, 75, 750, and 2250 ppm daily from gestation day 0 through lactation day 21. F(1) offspring were evaluated using the following tests: detailed clinical observations (postnatal days [PNDs] 4, 11, 21, 35, 45, and 60), auditory startle (PNDs 20 and 60), motor activity (PNDs 13, 17, 21, and 61), learning and memory using the Biel water maze (PNDs 22 and 62), and brain and nervous system neuropathology and brain morphometry (PNDs 21 and 72). For F(1) offspring, there were no treatment-related neurobehavioral effects, nor was there evidence of neuropathology or effects on brain morphometry. Based on maternal and offspring body weight reductions, the no-observed-adverse-effect level (NOAEL) for systemic toxicity was 75 ppm (5.85 and 13.1 mg/kg/day during gestation and lactation, respectively), with no treatment-related effects at lower doses or nonmonotonic dose responses observed for any parameter. There was no evidence that BPA is a developmental neurotoxicant in rats, and the NOAEL for developmental neurotoxicity was 2250 ppm, the highest dose tested (164 and 410 mg/kg/day during gestation and lactation, respectively).


Asunto(s)
Contaminantes Ocupacionales del Aire/toxicidad , Enfermedades del Sistema Nervioso/inducido químicamente , Sistema Nervioso/efectos de los fármacos , Fenoles/toxicidad , Anomalías Inducidas por Medicamentos , Animales , Animales Recién Nacidos , Compuestos de Bencidrilo , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Femenino , Lactancia/efectos de los fármacos , Longevidad/efectos de los fármacos , Masculino , Exposición Materna , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Sistema Nervioso/embriología , Sistema Nervioso/crecimiento & desarrollo , Enfermedades del Sistema Nervioso/embriología , Enfermedades del Sistema Nervioso/patología , Embarazo , Ratas , Ratas Sprague-Dawley
14.
Birth Defects Res B Dev Reprod Toxicol ; 86(6): 418-28, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20025038

RESUMEN

Assessment of potential developmental and reproductive toxicity of human pharmaceuticals is currently guided by the International Conference on Harmonization (ICH) S5(R2) document (available at http://www.ich.org). The studies that assess developmental hazard are generally conducted in rodents and rabbits. Based on the authors' collective experience, adequate designs (including range-finding studies) and the presentation of data for these studies are described in detail. In addition, the suggested initiation and then total duration of these studies in relation to clinical studies that enroll women of childbearing potential are described. Optional parameters that may be included in the studies are discussed, as are study designs that combine assessments of fertility and developmental toxicity. New methods that may replace or enhance current procedures are outlined. The details described herein will assist all laboratories performing these studies, individuals who need to plan for the studies, and regulatory agencies that ultimately review these studies.


Asunto(s)
Anomalías Inducidas por Medicamentos , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Pruebas de Toxicidad/métodos , Animales , Animales Endogámicos , Femenino , Agencias Internacionales , Cooperación Internacional , Masculino , Exposición Materna , Ratones , Conejos , Ratas , Proyectos de Investigación
15.
Birth Defects Res B Dev Reprod Toxicol ; 86(6): 429-36, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20025039

RESUMEN

Assessment of potential developmental and reproductive toxicity of human pharmaceuticals is currently guided by the ICH S5(R2) document, "Detection of Toxicity to Reproduction for Medicinal Products and Toxicity to Male Fertility." Studies that assess a candidate drug's effect on fertility are generally conducted in rats. The evolution of, and ultimate harmonization of, fertility study designs are reviewed, and specific elements of an acceptable design, as well as the recommendations for presentation of data, are described in detail. Additionally, the timing of nonclinical fertility studies in relation to clinical studies that enroll men and women of reproductive potential is reviewed. Possible strategies for combining fertility assessment with other study designs are also presented. This article provides testing laboratories, sponsors, and regulatory agencies with a comparison of current methods and designs, with the aim of providing a common understanding of the critical design features.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Fertilidad/efectos de los fármacos , Proyectos de Investigación , Pruebas de Toxicidad/métodos , Animales , Desarrollo Embrionario/efectos de los fármacos , Femenino , Agencias Internacionales , Cooperación Internacional , Masculino , Modelos Animales , Ratas , Medición de Riesgo , Factores de Tiempo
16.
Artículo en Inglés | MEDLINE | ID: mdl-20025047

RESUMEN

The objective of juvenile animal toxicity studies of pharmaceuticals is to obtain safety data, including information on the potential for adverse effects on postnatal growth and development. Studies in juvenile animals may assist in identifying postnatal developmental toxicities or other adverse effects that are not adequately assessed in the routine toxicity evaluations and that cannot be safely or adequately measured in pediatric clinical trials. Unlike the traditional reproductive and developmental toxicology studies that have been discussed in the accompanying reports, the design requirements for toxicity studies in juvenile animals are not explicitly defined in regulatory guidance. However, studies in juvenile animals can be useful in providing safety information necessary to enable pediatric clinical trials in pediatric patients or when there are special concerns for toxicities that cannot be safely or adequately measured in clinical trials. These juvenile animal toxicity studies are designed on a case-by-case basis. General design considerations and examples of study designs for assessment of juvenile animal toxicity are discussed.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Crecimiento y Desarrollo/efectos de los fármacos , Pediatría , Proyectos de Investigación , Pruebas de Toxicidad/métodos , Animales , Animales de Laboratorio , Animales Recién Nacidos , Femenino , Masculino , Preparaciones Farmacéuticas/clasificación , Ratas , Ratas Endogámicas
17.
Reprod Toxicol ; 27(3-4): 387-399, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19429409

RESUMEN

Perfluorooctanesulfonate (PFOS), a persistent and accumulative compound, is widely distributed in humans and wildlife. Human exposure can occur early in development, as evidenced by the detection of PFOS in umbilical cord blood and breast milk. As part of a developmental neurotoxicology study for which developmental endpoints, including those related to the developing nervous system, have been reported separately, groups of 25 pregnant Sprague Dawley rats were given daily oral doses of either vehicle control or potassium PFOS (K(+)PFOS) at 0.1, 0.3, and 1.0mg/kg-d from gestation day (GD) 0 (day positive for mating) through postnatal day (PND) 20. An additional 10 pregnant females per treatment group were treated through GD 19 and sacrificed on GD 20 in order to obtain maternal and fetal serum and tissue samples at the end of gestation. The present paper reports the results of samples of serum, liver, brain, and thyroid glands taken at various times to evaluate: (1) serum, liver, and brain PFOS concentrations by LC-MS/MS to establish the relationship between PFOS concentrations and study outcomes; (2) serum thyrotropin (TSH) concentrations by RIA; (3) thyroid follicular cell proliferation index by Ki-67 immunohistochemical staining; (4) thyroid follicle epithelial cell height and colloidal area by histomorphometric analysis; (5) selected liver mRNA transcripts by quantitative RT-PCR. PFOS concentrations in dam and pup serum, liver, and brain increased across treatment groups in approximate proportion to the proportional increases in maternal K(+)PFOS dose, and sex differences in PFOS concentrations were not apparent in pups on PND 21. In pups from K(+)PFOS maternal dose groups on PND 72, serum PFOS had decreased to about 3 and 11% of PND 21 concentrations in males and females, respectively, and liver PFOS had decreased to about 17% of PND 21 concentrations in both sexes. Liver PFOS concentrations were approximately 0.6-0.8 times serum PFOS in GD 20 fetuses, and increased to about 2-4 times serum concentrations on PND 4 and 21. GD 20 fetal and PND 4 pup brain PFOS concentrations were approximately 33% of the corresponding serum concentrations, dropping to approximately 10% by PND 21, in contrast to dam brain PFOS concentrations, which were approximately 4-9% of serum PFOS concentrations. Compared to controls, Cyp2b2 mRNA was increased (2.8-fold) in the 1.0mg/kg-d treatment-group dams on GD 20. In male pups on PND 21, Cyp4A1, ACoA, and Cyp2b2 were increased 2.1-, 1.5-, and 1.8-fold, respectively, and Cyp7A1 was decreased 3.5-fold. Serum TSH and thyroid follicular morphology were not altered by K(+)PFOS treatment. The mean number of proliferating thyroid follicular cells was increased 2.1-fold over control in GD 20 female fetuses from 1.0mg/kg-d-treated dams, yet the highest individual count was similar to that of controls (116 versus 113 in controls).


Asunto(s)
Ácidos Alcanesulfónicos/farmacocinética , Contaminantes Ambientales/farmacocinética , Fluorocarburos/farmacocinética , Expresión Génica/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Hormonas Tiroideas/farmacocinética , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/sangre , Ácidos Alcanesulfónicos/farmacología , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/sangre , Contaminantes Ambientales/farmacología , Femenino , Fluorocarburos/administración & dosificación , Fluorocarburos/sangre , Fluorocarburos/farmacología , Edad Gestacional , Lactancia , Masculino , Exposición Materna , Embarazo , Ratas , Ratas Sprague-Dawley , Hormonas Tiroideas/sangre , Tiroxina/sangre , Tiroxina/farmacocinética , Triyodotironina/sangre , Triyodotironina/farmacocinética
18.
Food Chem Toxicol ; 47(6): 1296-303, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19285533

RESUMEN

Evidence suggests that the health benefits associated with green tea consumption are related to tea catechins. The objective of this study was to evaluate potential maternal and fetal effects of standardized heat-sterilized green tea catechins (GTC-H). GTC-H was gavage administered to mated female rats from gestation day 6 through 17, at doses of 0, 200, 600, and 2000 mg/kg/day. There were no GTC-H-related deaths or macroscopic findings. During the entire gestation period in the high-dose (2000 mg/kg/day)-treated group and during days 6-9 and 6-18 in the 600 mg/kg/day group, mean body weight gain was lower. Mean feed consumption was lower during gestation days 6-9 in the 600 mg/kg/day group and during gestation days 6-9 and 9-12 in the 2000 mg/kg/day group. Compared to the control group, mean body weights in the 600 and 2000 mg/kg/day groups were up to 5.1% and 7.7% lower during gestation days 9-20. GTC-H administration did not affect mean gravid uterine weights or intrauterine growth and survival. There were no GTC-H-related fetal malformations or developmental variations. Based on the results of this study, the no-observed-adverse-effect level (NOAEL) for GTC-H was 200mg/kg/day for maternal toxicity, and 2000 mg/kg/day for embryo/fetal development.


Asunto(s)
Catequina/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Té/química , Animales , Peso Corporal/efectos de los fármacos , Química Farmacéutica , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Feto/patología , Histerectomía , Masculino , Nivel sin Efectos Adversos Observados , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Sobrevida , Útero/patología
19.
Reprod Toxicol ; 27(3-4): 319-330, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19162172

RESUMEN

Perfluorooctanesulfonate (PFOS), a persistent and bioaccumulative compound, is widely distributed in humans and wildlife. Exposure of the human fetus and neonate to PFOS can occur in utero and via the mother's milk, respectively. Developmental studies have been conducted with PFOS in the past, including some developmental neurotoxicity endpoints. The objective of this study was to evaluate the functional and morphological changes to the nervous system in rats having gestational and lactational exposures to PFOS per current test guidelines (EPA OPPTS 870.6300 and OECD 426). Female SD rats (25/dosage group) were given daily oral doses of either 0.0, 0.1, 0.3, or 1.0mg/kg-d potassium PFOS (K(+)PFOS) from gestation day (GD) 0 through postnatal day (PND) 20. Offspring were observed through PND 72 for growth, maturation, motor activity, learning and memory, acoustic startle reflex, various behavioral manifestations, and brain weight. Specimens were taken from dams, fetuses, and pups for serum and tissue PFOS concentration, thyroid status endpoints, and liver mRNA transcript analysis, and those results are reported in a companion article. No significant effect was noted on maternal health or reproductive outcomes from dosing of maternal rats with K(+)PFOS throughout gestation. Maternal body weights were statistically significantly lower in the 1.0mg/kg-d dosage group from PND 4 through the end of lactation. Offspring from K(+)PFOS-treated maternal groups did not differ significantly from controls with respect to birth weight, growth, age and weight at attainment of sexual maturation, learning and memory, acoustic startle, various behavioral endpoints, and brain weight. Male offspring from the 1.0mg/kg-d maternal treatment group displayed increased motor activity and reduced habituation on PND 17 but not on PND 13, 21, and 61. The maternal no-observed-adverse-effect-level (NOAEL) was 0.3mg/kg-d based on decreased body weights observed in lactation. The maternal dose associated with the NOAEL for male offspring was 0.3mg/kg-d based on increased motor activity and reduced habituation in the 1.0mg/kg-d maternal dose-group male offspring on PND 17. The maternal dose associated with the NOAEL for female offspring was >1.0mg/kg-d. Mean serum concentrations of PFOS reported in a companion article for the 0.3mg/kg-d group maternal rats are several hundred times higher than those reported for females in the United States general population.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Lactancia/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Efectos Tardíos de la Exposición Prenatal , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Edad Gestacional , Guías como Asunto , Exposición Materna , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Embarazo , Distribución Aleatoria , Ratas , Ratas Endogámicas , Reproducción/efectos de los fármacos
20.
Food Chem Toxicol ; 47(4): 681-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19059452

RESUMEN

SLE66 a synthetic pseudo-ceramide, has been shown to reduce dryness/scaling/itching of human skin. Naturally occurring ceramides have been claimed to play a crucial role in cell proliferation, differentiation, and apoptosis including processes important for embryogenesis. The objective of this study was to evaluate the potential maternal and fetal effects of SLE66. SLE66 was administered orally (gavage) to mated female Crl:CD(SD)IGS BR rats (25/group) once daily from gestation day 6 through 19, at dose levels of 0 (control), 150, 400 or 1000 mg/kg/day. No treatment-related clinical or internal (macroscopic) findings were noted and all animals survived to the scheduled necropsy on gestation day 20. SLE66 administration did not affect mean maternal body weights, body weight gains, net body weights, net body weight gains, gravid uterine weights, or feed consumption. Similarly, SLE66 administration did not affect intrauterine growth and survival related parameters such as viable fetuses, pre-implantation loss, early and late resorptions, fetal weight and fetal sex. No SLE66-related fetal malformations or developmental variations were noted. Based on the results of this study, a dose level of 1000 mg/kg/day (highest dose used) was considered as the no-observed-adverse-effect level (NOAEL) for both maternal and developmental toxicity.


Asunto(s)
Anomalías Inducidas por Medicamentos , Ceramidas/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Nivel sin Efectos Adversos Observados , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...