Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Chem Res Toxicol ; 36(3): 380-389, 2023 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-36821091

RESUMEN

Over the past decade, there has been increased concern for environmental chemicals that can target various sites within the hypothalamic-pituitary-thyroid axis to potentially disrupt thyroid synthesis, transport, metabolism, and/or function. One well-known thyroid target in both humans and wildlife is the sodium iodide symporter (NIS) that regulates iodide uptake into the thyroid gland, the first step of thyroid hormone synthesis. Our laboratory previously developed and validated a radioactive iodide uptake (RAIU) high-throughput assay in a stably transduced human NIS cell line (hNIS-HEK293T-EPA) to identify chemicals with potential for NIS inhibition. So far, we have tested over 2000 chemicals (US EPA's ToxCast chemical libraries PI_v2, PII, and e1K) and discovered a subset of chemicals that significantly inhibit iodide uptake in the hNIS assay. Here, we utilized this screening assay to test a set of 149 unique per- and polyfluoroalkyl substances (PFAS) (ToxCast PFAS library) for potential NIS inhibition. For this evaluation, the 149 blinded samples were screened in a tiered approach, first in an initial single-concentration (≤100 µM) RAIU assay and subsequent evaluation of the chemicals that produced ≥20% inhibition using multiconcentration (MC) response (0.001-100 µM) testing in parallel RAIU and cell viability assays. Of this set, 38 of the PFAS chemicals inhibited iodide uptake ≥20% in the MC testing with 25 displaying inhibition ≥50%. To prioritize the most potent PFAS NIS inhibitors in this set, chemicals were ranked based on outcomes of both iodide uptake and cytotoxicity and normalized to perchlorate, a known positive control. Consistent with previous findings, PFOS and PFHxS were again found to be potent NIS inhibitors, yet significant inhibition was also observed for several other screened PFAS chemicals. Although further studies are clearly warranted, this initial screening effort identifies NIS as a molecular target for potential thyroid disruption by this persistent and structurally diverse class of chemicals.


Asunto(s)
Fluorocarburos , Ensayos Analíticos de Alto Rendimiento , Humanos , Bibliotecas de Moléculas Pequeñas/toxicidad , Yoduros/farmacología , Yoduros/metabolismo , Células HEK293
2.
Arch Toxicol ; 95(5): 1723-1737, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33656581

RESUMEN

The sodium-iodide symporter (NIS) mediates the uptake of iodide into the thyroid. Inhibition of NIS function by xenobiotics has been demonstrated to suppress circulating thyroid hormones and perturb related physiological functions. Until recently, few environmental chemicals had been screened for NIS inhibition activity. We previously screened over 1000 chemicals from the ToxCast Phase II (ph1v2 and ph2) libraries using an in vitro radioactive iodide uptake (RAIU) with the hNIS-HEK293T cell line to identify NIS inhibitors. Here, we broaden the chemical space by expanding screening to include the ToxCast e1k library (804 unique chemicals) with initial screening for RAIU at 1 × 10-4 M. Then 209 chemicals demonstrating > 20% RAIU inhibition were further tested in multiple-concentration, parallel RAIU and cell viability assays. This identified 55 chemicals as active, noncytotoxic RAIU inhibitors. Further cytotoxicity-adjusted potency scoring (with NaClO4 having a reference score of 200) revealed five chemicals with moderate to strong RAIU inhibition (scored > 100). These data were combined with our previous PhII screening data to produce binary hit-calls for ~ 1800 unique chemicals (PhII + e1k) with and without cytotoxicity filtering. Results were analyzed with a ToxPrint chemotype-enrichment workflow to identify substructural features significantly enriched in the NIS inhibition hit-call space. We assessed the applicability of enriched PhII chemotypes to prospectively predict NIS inhibition in the e1k dataset. Chemotype enrichments derived for the combined ~ 1800 dataset also identified additional enriched features, as well as chemotypes affiliated with cytotoxicity. These enriched chemotypes provide important new information that can support future data interpretation, structure-activity relationship, chemical use, and regulation.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Simportadores/antagonistas & inhibidores , Animales , Bioensayo , Transporte Biológico , Supervivencia Celular , Células HEK293 , Humanos , Yoduros , Relación Estructura-Actividad , Glándula Tiroides
3.
Arch Toxicol ; 94(3): 873-885, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32065294

RESUMEN

The Fischer rat thyroid follicular cell line (FRTL-5) endogenously expresses the sodium-iodide symporter (NIS) and has been used to identify environmental chemicals that perturb thyroid hormone homeostasis by disruption of NIS-mediated iodide uptake. Previously, a high-throughput radioactive iodide uptake (RAIU) screening assay incorporating the hNIS-HEK293T-EPA cell line was used to identify potential human NIS (hNIS) inhibitors in 1028 ToxCast Phase I (ph1_v2) and Phase II chemicals. In this study, the FRTL-5 cell line was evaluated and applied as a secondary RAIU assay coupled with cell viability assays to further prioritize highly active NIS inhibitors from the earlier screening. Assay validation with ten reference chemicals and performance assessment by chemical controls suggest the FRTL-5 based assays are robust and highly reproducible. Top-ranked chemicals from the ToxCast screening were then evaluated in both FRTL-5 and hNIS RAIU assays using newly sourced chemicals to strengthen the testing paradigm and to enable a rat vs. human species comparison. Eighteen of 29 test chemicals showed less than 1 order of magnitude difference in IC50 values between the two assays. Notably, two common perfluorinated compounds, perfluorooctanesulfonic acid (PFOS) and perfluorohexane sulfonate (PFHxS), demonstrated strong NIS inhibitory activity [IC50 - 6.45 (PFOS) and - 5.70 (PFHxS) log M in FRTL-5 RAIU assay]. In addition, several chemicals including etoxazole, methoxyfenozide, oxyfluorfen, triclocarban, mepanipyrim, and niclosamide also exhibited NIS inhibition with minimal cytotoxicity in both assays and are proposed for additional testing using short-term in vivo assays to characterize effects on thyroid hormone synthesis.


Asunto(s)
Yoduros/metabolismo , Simportadores/metabolismo , Animales , Bioensayo , Transporte Biológico , Humanos , Ratas , Ratas Endogámicas F344 , Simportadores/antagonistas & inhibidores , Células Epiteliales Tiroideas
4.
Environ Sci Technol ; 52(9): 5417-5426, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29611697

RESUMEN

Thyroid uptake of iodide via the sodium-iodide symporter (NIS) is the first step in the biosynthesis of thyroid hormones that are critical for health and development in humans and wildlife. Despite having long been a known target of endocrine disrupting chemicals such as perchlorate, information regarding NIS inhibition activity is still unavailable for the vast majority of environmental chemicals. This study applied a previously validated high-throughput approach to screen for NIS inhibitors in the ToxCast phase I library, representing 293 important environmental chemicals. Here 310 blinded samples were screened in a tiered-approach using an initial single-concentration (100 µM) radioactive-iodide uptake (RAIU) assay, followed by 169 samples further evaluated in multi-concentration (0.001 µM-100 µM) testing in parallel RAIU and cell viability assays. A novel chemical ranking system that incorporates multi-concentration RAIU and cytotoxicity responses was also developed as a standardized method for chemical prioritization in current and future screenings. Representative chemical responses and thyroid effects of high-ranking chemicals are further discussed. This study significantly expands current knowledge of NIS inhibition potential in environmental chemicals and provides critical support to U.S. EPA's Endocrine Disruptor Screening Program (EDSP) initiative to expand coverage of thyroid molecular targets, as well as the development of thyroid adverse outcome pathways (AOPs).


Asunto(s)
Disruptores Endocrinos , Simportadores , Humanos , Yoduros , Glándula Tiroides
5.
J Toxicol Environ Health A ; 80(4): 236-249, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28569618

RESUMEN

Triclosan (TCS), an antibacterial, has been shown to be an endocrine disruptor in the rat. Previously, subchronic TCS treatment to female rats was found to advance puberty and potentiate the effect of ethinyl estradiol (EE) on uterine growth when EE and TCS were co-administered prior to weaning. In the pubertal study, a decrease in serum thyroxine (T4) concentrations with no significant change in serum thyroid-stimulating hormone (TSH) was also observed. The purpose of the present study was to further characterize the influence of TCS on the reproductive and thyroid axes of the female rat using a chronic exposure regimen. Female Wistar rats were exposed by oral gavage to vehicle control, EE (1 µg/kg), or TCS (2.35, 4.69, 9.375 or 37.5 mg/kg) for 8 months and estrous cyclicity monitored. Although a divergent pattern of reproductive senescence appeared to emerge from 5 to 11 months of age between controls and EE-treated females, no significant difference in cyclicity was noted between TCS-treated and control females. A higher % control females displayed persistent diestrus (PD) by the end of the study, whereas animals administered with positive control (EE) were predominately persistent estrus (PE). Thyroxine concentration was significantly decreased in TCS-administered 9.375 and 37.5 mg/kg groups, with no marked effects on TSH levels, thyroid tissue weight, or histology. Results demonstrate that a long-term exposure to TCS did not significantly alter estrous cyclicity or timing of reproductive senescence in females but suppressed T4 levels at a lower dose than previously observed.


Asunto(s)
Envejecimiento/efectos de los fármacos , Ciclo Estral/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Reproducción/efectos de los fármacos , Glándula Tiroides/efectos de los fármacos , Triclosán/toxicidad , Animales , Antiinfecciosos Locales/toxicidad , Femenino , Ratas , Ratas Wistar , Pruebas de Toxicidad Crónica
6.
Toxicol Sci ; 198(1): 113-127, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38145495

RESUMEN

The environmental contaminant perchlorate impairs the synthesis of thyroid hormones by reducing iodine uptake into the thyroid gland. Despite this known action, moderate doses of perchlorate do not significantly alter serum thyroid hormone in rat pups born to exposed dams. We examined perchlorate dosimetry and responsivity of the thyroid gland and brain in offspring following maternal exposure to perchlorate. Pregnant rat dams were delivered perchlorate in drinking water (0, 30, 100, 300, 1000 ppm) from gestational day 6 to postnatal day (PN) 21. Perchlorate was present in the placenta, milk, and serum, the latter declining in pups over the course of lactation. Serum and brain thyroid hormone were reduced in pups at birth but recovered to control levels by PN2. Dramatic upregulation of Nis was observed in the thyroid gland of the exposed pup. Despite the return of serum thyroid hormone to control levels by PN2, expression of several TH-responsive genes was altered in the PN14 pup brain. Contextual fear learning was unimpaired in the adults, supporting previous reports. Declining levels of serum perchlorate and a profound upregulation of Nis gene expression in the thyroid gland are consistent with the rapid return to the euthyroid state in the neonate. However, despite this recovery, thyroid hormone insufficiencies in serum and brain beginning in utero and present at birth appear sufficient to alter TH action in the fetus and subsequent trajectory of brain development. Biomarkers of that altered trajectory remain in the brain of the neonate, demonstrating that perchlorate is not devoid of effects on the developing brain.


Asunto(s)
Compuestos de Amonio Cuaternario , Resiliencia Psicológica , Glándula Tiroides , Embarazo , Femenino , Ratas , Animales , Percloratos/toxicidad , Percloratos/metabolismo , Animales Recién Nacidos , Hormonas Tiroideas
7.
Birth Defects Res B Dev Reprod Toxicol ; 98(5): 428-35, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24323408

RESUMEN

Two critical components of the validation of any in vivo screening assay are to demonstrate sensitivity and specificity. Although the Endocrine Disruptor Screening Program's Tier 1 Male and Female Pubertal Protocols have been shown to be sensitive assays for the detection of weak endocrine disrupting chemicals (EDCs), there are concerns that the assays lack specificity for EDC effects when a chemical induces systemic toxicity. A lack of specificity, or the ability to correctly identify an inactive or "negative" chemical, would increase the probability of identifying false positives. Here, we orally exposed rats to hydroxyatrazine (OH-ATR), a biotransformation by-product of the chlorotriazine herbicides that produced nephrotoxicity following a 13-week dietary exposure. Based on a previous study in our laboratory, males were dosed with 11.4 to 183.4 mg/kg OH-ATR and females were dosed with 45.75 to 183.4 mg/kg OH-ATR. Following exposure in both sexes, there was a dose-response increase in mean kidney weights and the incidence and severity of kidney lesions. These lesions included the deposition of mineralized renal tubule concretions, hydronephrosis, renal tubule dilatation, and pyelonephritis. However, no differences in body weight, liver weight, or reproductive tissue weights, reproductive or thyroid histology, hormone concentrations or the age of pubertal onset were observed. Therefore, the results demonstrate that the endpoints included in the pubertal assay are useful for nonendocrine (systemic) effects that define an no observable effect level (NOEL) or lowest observable effect level (LOEL) and provide one example where an impact on kidney function does not alter any of the endocrine-specific endpoints of the assay.


Asunto(s)
Atrazina/análogos & derivados , Disruptores Endocrinos/toxicidad , Maduración Sexual/efectos de los fármacos , Animales , Atrazina/toxicidad , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Wistar
8.
Neurotoxicol Teratol ; 100: 107303, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37777095

RESUMEN

Identifying xenobiotics that interrupt the thyroid axis has significant public health implications, given that thyroid hormones are required for brain development. As such, some developmental and reproductive toxicology (DART) studies now require or recommend serum total thyroxine (T4) measurements in pregnant, lactating, and developing rats. However, serum T4 concentrations are normally low in the fetus and pup which makes quantification difficult. These challenges can be circumvented by technologies like mass spectrometry, but these approaches are expensive and not always widely available. To demonstrate the feasibility of measuring T4 using a commercially available assay, we examine technical replicates of rat serum samples measured both by liquid chromatography mass spectrometry (LC/MS/MS) and radioimmunoassay (RIA). These samples were obtained from rats on gestational day 20 (dams and fetuses) or postnatal day 5 (pups), following maternal exposure to the goitrogen propylthiouracil (0-3 ppm) to incrementally decrease T4. We show that with assay modification, it is possible to measure serum T4 using low sample volumes (25-50 µL) by an RIA, including in the GD20 fetus exposed to propylthiouracil. This proof-of-concept study demonstrates the technical feasibility of measuring serum T4 in DART studies.


Asunto(s)
Tiroxina , Triyodotironina , Embarazo , Femenino , Ratas , Animales , Propiltiouracilo , Radioinmunoensayo/métodos , Espectrometría de Masas en Tándem/métodos , Lactancia , Feto
9.
Front Endocrinol (Lausanne) ; 14: 1090081, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36843608

RESUMEN

Thyroid hormone (TH) action controls brain development in a spatiotemporal manner. Previously, we demonstrated that perinatal hypothyroidism led to formation of a periventricular heterotopia in developing rats. This heterotopia occurs in the posterior telencephalon, and its formation was preceded by loss of radial glia cell polarity. As radial glia mediate cell migration and originate in a progenitor cell niche called the ventricular zone (VZ), we hypothesized that TH action may control cell signaling in this region. Here we addressed this hypothesis by employing laser capture microdissection and RNA-Seq to evaluate the VZ during a known period of TH sensitivity. Pregnant rats were exposed to a low dose of propylthiouracil (PTU, 0.0003%) through the drinking water during pregnancy and lactation. Dam and pup THs were quantified postnatally and RNA-Seq of the VZ performed in neonates. The PTU exposure resulted in a modest increase in maternal thyroid stimulating hormone and reduced thyroxine (T4). Exposed neonates exhibited hypothyroidism and T4 and triiodothyronine (T3) were also reduced in the telencephalon. RNA-Seq identified 358 differentially expressed genes in microdissected VZ cells of hypothyroid neonates as compared to controls (q-values ≤0.05). Pathway analyses showed processes like maintenance of the extracellular matrix and cytoskeleton, cell adhesion, and cell migration were significantly affected by hypothyroidism. Immunofluorescence also demonstrated that collagen IV, F-actin, radial glia, and adhesion proteins were reduced in the VZ. Immunohistochemistry of integrin αvß3 and isoforms of both thyroid receptors (TRα/TRß) showed highly overlapping expression patterns, including enrichment in the VZ. Taken together, our results show that TH action targets multiple components of cell junctions in the VZ, and this may be mediated by both genomic and nongenomic mechanisms. Surprisingly, this work also suggests that the blood-brain and blood-cerebrospinal fluid barriers may also be affected in hypothyroid newborns.


Asunto(s)
Hipotiroidismo , Tiroxina , Embarazo , Femenino , Ratas , Animales , Animales Recién Nacidos , Tiroxina/metabolismo , Antitiroideos , Hormonas Tiroideas/metabolismo , Hipotiroidismo/metabolismo , Encéfalo/metabolismo , Uniones Intercelulares/metabolismo
10.
Environ Int ; 126: 377-386, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30826616

RESUMEN

In support of the Endocrine Disruptor Screening Program (EDSP), the U.S.EPA's Office of Research and Development (ORD) is developing high-throughput screening (HTS) approaches to identify chemicals that alter target sites in the thyroid hormone (TH) pathway. The sodium iodide symporter (NIS) is a transmembrane glycoprotein that mediates iodide uptake into the thyroid as the initial step of TH biosynthesis. Previously, we screened 293 ToxCast chemicals (ph1v2) using a HEK293T cell line expressing human NIS in parallel radioactive iodide uptake (RAIU) and cell viability assays to identify potential environmental NIS inhibitors. Here, we expanded NIS inhibitor screening for a set of 768 ToxCast Phase II (ph2) chemicals, and applied a novel computational toxicology approach based on the ToxPrint chemotype to identify chemical substructures associated with NIS inhibition. Following single-concentration screening (at 1 × 10-4 M with a 20% inhibition cutoff), 235 samples (228 chemicals) were further tested in multiple-concentration (1 × 10-9 - 1 × 10-4 M) format in both RAIU and cell viability assays. The 167 chemicals that exhibited significant RAIU inhibition were then prioritized using combined RAIU and cell viability responses that were normalized relative to the known NIS inhibitor sodium perchlorate. Some of the highest ranked chemicals, such as PFOS, tributyltin chloride, and triclocarban, have been previously reported to be thyroid disruptors. In addition, several novel chemicals were identified as potent NIS inhibitors. The present results were combined with the previous ph1v2 screening results to produce two sets of binary hit-calls for 1028 unique chemicals, consisting of 273 positives exhibiting significant RAIU inhibition, and 63 positives following application of a cell viability filter. A ToxPrint chemotype-enrichment analysis identified >20 distinct chemical substructural features, represented in >60% of the active chemicals, as significantly enriched in each NIS inhibition hit-call space. A shared set of 9 chemotypes enriched in both hit-call sets indicates stable chemotype signals (insensitive to cytotoxicity filters) that can help guide structure-activity relationship (SAR) investigations and inform future research.


Asunto(s)
Disruptores Endocrinos/toxicidad , Ensayos Analíticos de Alto Rendimiento , Simportadores/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Humanos
11.
Environ Health Perspect ; 127(9): 95001, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31487205

RESUMEN

BACKGROUND: Extensive clinical and experimental research documents the potential for chemical disruption of thyroid hormone (TH) signaling through multiple molecular targets. Perturbation of TH signaling can lead to abnormal brain development, cognitive impairments, and other adverse outcomes in humans and wildlife. To increase chemical safety screening efficiency and reduce vertebrate animal testing, in vitro assays that identify chemical interactions with molecular targets of the thyroid system have been developed and implemented. OBJECTIVES: We present an adverse outcome pathway (AOP) network to link data derived from in vitro assays that measure chemical interactions with thyroid molecular targets to downstream events and adverse outcomes traditionally derived from in vivo testing. We examine the role of new in vitro technologies, in the context of the AOP network, in facilitating consideration of several important regulatory and biological challenges in characterizing chemicals that exert effects through a thyroid mechanism. DISCUSSION: There is a substantial body of knowledge describing chemical effects on molecular and physiological regulation of TH signaling and associated adverse outcomes. Until recently, few alternative nonanimal assays were available to interrogate chemical effects on TH signaling. With the development of these new tools, screening large libraries of chemicals for interactions with molecular targets of the thyroid is now possible. Measuring early chemical interactions with targets in the thyroid pathway provides a means of linking adverse outcomes, which may be influenced by many biological processes, to a thyroid mechanism. However, the use of in vitro assays beyond chemical screening is complicated by continuing limits in our knowledge of TH signaling in important life stages and tissues, such as during fetal brain development. Nonetheless, the thyroid AOP network provides an ideal tool for defining causal linkages of a chemical exerting thyroid-dependent effects and identifying research needs to quantify these effects in support of regulatory decision making. https://doi.org/10.1289/EHP5297.


Asunto(s)
Rutas de Resultados Adversos , Contaminantes Ambientales/toxicidad , Glándula Tiroides/efectos de los fármacos , Animales , Bioensayo , Humanos , Hormonas Tiroideas
12.
Toxicol Sci ; 100(1): 194-202, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17728285

RESUMEN

The U.S. Environmental Protection Agency is currently validating assays that will be used in a Tier I Screening Battery to detect endocrine disrupting chemicals. A primary concern with the Protocols for the Assessment of Pubertal Development and Thyroid Function in Juvenile Male and Female Rats is that a nonspecific reduction in body weight (BWT) during the exposure period may potentially confound the interpretation of effects on the endocrine endpoints. Wistar rats were underfed 10, 20, 30, or 40% less than the ad libitum food consumed by controls from postnatal days (PNDs) 22 to 42 (females) or PNDs 23 to 53 (males). Terminal BWT of females and males were 2, 4, 12, and 19% and 2, 6, 9, and 19% lower than controls, respectively. In the females, neither the age of pubertal onset nor any of the thyroid hormone endpoints were affected by food restriction (FR) that led to a 12% decrease in BWT. Similarly, none of the male reproductive endpoints examined were altered by FR that led to a 9% BWT decrease. However, decreased triiodothyronine and thyroxin was observed in FR males with a 9% reduced BWT. While these data support the use of the maximum tolerated dose for BWT (10%) for the female protocol, effects on the male thyroid endpoints indicate that a slightly lower limit (

Asunto(s)
Peso Corporal/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Privación de Alimentos , Maduración Sexual , Pruebas de Toxicidad/métodos , Pérdida de Peso , Glándulas Suprarrenales/crecimiento & desarrollo , Animales , Glucemia/metabolismo , Femenino , Genitales Femeninos/crecimiento & desarrollo , Genitales Masculinos/crecimiento & desarrollo , Riñón/crecimiento & desarrollo , Leptina/sangre , Hígado/crecimiento & desarrollo , Masculino , Tamaño de los Órganos , Hipófisis/crecimiento & desarrollo , Ratas , Ratas Wistar , Reproducibilidad de los Resultados , Glándula Tiroides/crecimiento & desarrollo , Hormonas Tiroideas/sangre , Pruebas de Toxicidad/normas , Vagina/crecimiento & desarrollo
13.
Reprod Toxicol ; 23(4): 607-10, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17399945

RESUMEN

The purpose of the present study was to examine the distribution of atrazine in the lactating dam and suckling neonate following an acute exposure to either 2 or 4mg/kg 14C-atrazine (14C-ATR) by gavage. 14C-ATR was administered to the nursing dam on postnatal day 3 by oral gavage. Two and a half hours after exposure of the mother to 14C-ATR, the pups were allowed to nurse for 30min. At the end of the nursing period, radiolabelled residues of 14C-ATR [or 14C-chlorotriazines (14C-ClTRI)] were measured in the organs and tissues of the perfused dam and in the stomachs and brains of the rat pups. Both the 2 and the 4mg atrazine treatments resulted in a transfer of approximately 0.007% of 14C-ClTRI to the stomach (indicator of milk content) and 0.0002% to the brains of the offspring following the 30-min nursing period. Three hours following the dose of 14C-ATR, there was a distribution of 14C-ClTRI to the organs of the dam, with the highest amounts in the liver and kidney (1.1 and 0.3% of the administered dose, respectively). Approximately 0.003% of the administered dose was present in three different brain sections of the dam following both doses of 14C-ATR. The results of this study demonstrate that 14C-ClTRI are present in small concentrations in the brain and tissues of the dam (adult female) and provide evidence that atrazine or the metabolites can have direct effects on neuroendrocrine function. The results also provide information for postnatal distribution into the suckling neonate during early lactation.


Asunto(s)
Atrazina/farmacocinética , Herbicidas/farmacocinética , Lactancia , Administración Oral , Animales , Animales Lactantes , Atrazina/administración & dosificación , Atrazina/sangre , Radioisótopos de Carbono/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Herbicidas/administración & dosificación , Herbicidas/sangre , Nivel sin Efectos Adversos Observados , Ratas , Ratas Wistar , Medición de Riesgo , Distribución Tisular
14.
Toxicol In Vitro ; 40: 66-78, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27979590

RESUMEN

The U.S. EPA's Endocrine Disruptor Screening Program aims to use high-throughput assays and computational toxicology models to screen and prioritize chemicals that may disrupt the thyroid signaling pathway. Thyroid hormone biosynthesis requires active iodide uptake mediated by the sodium/iodide symporter (NIS). Monovalent anions, such as the environmental contaminant perchlorate, are competitive inhibitors of NIS, yet limited information exists for more structurally diverse chemicals. A novel cell line expressing human NIS, hNIS-HEK293T-EPA, was used in a radioactive iodide uptake (RAIU) assay to identify inhibitors of NIS-mediated iodide uptake. The RAIU assay was optimized and performance evaluated with 12 reference chemicals comprising known NIS inhibitors and inactive compounds. An additional 39 chemicals including environmental contaminants were evaluated, with 28 inhibiting RAIU over 20% of that observed for solvent controls. Cell viability assays were performed to assess any confounding effects of cytotoxicity. RAIU and cytotoxic responses were used to calculate selectivity scores to group chemicals based on their potential to affect NIS. RAIU IC50 values were also determined for chemicals that displayed concentration-dependent inhibition of RAIU (≥50%) without cytotoxicity. Strong assay performance and highly reproducible results support the utilization of this approach to screen large chemical libraries for inhibitors of NIS-mediated iodide uptake.


Asunto(s)
Disruptores Endocrinos/toxicidad , Yoduros/metabolismo , Simportadores/antagonistas & inhibidores , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Radioisótopos de Yodo , Simportadores/genética , Glándula Tiroides/metabolismo
15.
Toxicol Sci ; 84(1): 38-48, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15601673

RESUMEN

Molinate, a thiocarbamate herbicide, has been reported to impair reproductive capability in the male rat and alter pregnancy outcome in a two-generation study. Published data are lacking on the effects of acute exposure to molinate in the female. Based on this work and our previous observations with related dithiocarbamate compounds, we hypothesized that a single exposure to molinate during the critical window for the neural trigger of ovulation on the day of proestrus (PRO) would block the luteinizing hormone (LH) surge and delay ovulation. To examine the effect of molinate on the LH surge, ovariectomized (OVX) rats were implanted with Silastic capsules containing estradiol benzoate to mimic physiological levels on proestrus. Doses of 25 and 50 mg/kg molinate significantly suppressed LH and prolactin secretion. Intact regularly cycling females gavaged with 0, 25, or 50 mg/kg molinate at 1300 h on PRO were examined on estrus or estrus +1 day for the presence of oocytes in the oviduct. All control females had oocytes in the oviduct on estrus. Molinate doses of 6.25 to 50 mg/kg delayed ovulation for 24 h. Estrous cyclicity was examined after daily exposure to 50 mg/kg (21 days). Estrous cyclicity was irregular in the molinate group, showing extended days in estrus. Two experiments were conducted to determine whether molinate blocked the LH surge via a central nervous system (CNS) mode of action or via an alteration in pituitary response. In the first experiment, we evaluated the release of LH in control and molinate-treated rats after a bolus dose of exogenous GnRH. Luteinizing hormone release was comparable in the two groups, suggesting that the effect of molinate is centrally mediated. To further examine the potential role of the CNS, we examined the pulsatile release of LH present in the long-term OVX females. In this model, the pulsatile pattern of LH secretion is directly correlated with GnRH release from the hypothalamus. A significant decrease in the LH pulse frequency was observed in molinate-treated females. These results indicate that molinate is able to delay ovulation by suppressing the LH surge on the day of proestrus and that the brain is the primary target site for the effects on pituitary hormone secretion.


Asunto(s)
Azepinas/toxicidad , Herbicidas/toxicidad , Sistemas Neurosecretores/efectos de los fármacos , Ovulación/efectos de los fármacos , Tiocarbamatos/toxicidad , Animales , Encéfalo/enzimología , Colinesterasas/sangre , Colinesterasas/metabolismo , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Implantes de Medicamentos , Estrógenos/administración & dosificación , Estrógenos/farmacología , Ciclo Estral/efectos de los fármacos , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Norepinefrina/metabolismo , Ovariectomía , Hipófisis/metabolismo , Embarazo , Prolactina/sangre , Ratas , Ratas Long-Evans , Factores de Tiempo
16.
Toxicol Sci ; 67(2): 198-206, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12011479

RESUMEN

Recently we reported that atrazine (ATR), a chlorotriazine herbicide, alters the onset of puberty in male Wistar rats. In this study, we examined the same reproductive parameters in the developing male rat following a similar exposure to the primary, chlorinated metabolites of atrazine. Intact male Wistar rats were gavaged from postnatal day (PND) 23 through PND 53 and several reproductive endpoints were examined. The doses selected were the molar equivalents to atrazine in our previous work. Deethylatrazine (DEA), deisopropyl-atrazine (DIA), and diaminochlorotriazine (DACT) were administered by gavage at doses equivalent to the atrazine equimolar doses (AED) of 6.25, 12.5, 25, 50, 100, or 200 mg/kg. Preputial separation (PPS) was significantly delayed by DEA at 25, 100, and 200 AED, by DIA at 25, 100, and 200 AED, and by DACT at 12.5 through 200 AED. When the males were killed on PND 53, DEA (100 and 200 AED), DIA (50 through 200 AED), and DACT (200 AED) treatments caused a significant reduction in ventral prostate weight, while only the highest doses of DIA and DEA resulted in a significant decrease in lateral prostate weight. Seminal vesicle weight was reduced by DEA (25, 100, and 200 AED), DIA (100 and 200 AED), and 100 and 200 AED of DACT. Epididymal weights were reduced in the DEA (200 AED), DIA (200 AED), and DACT (100 and 200 AED) treatment groups. Serum testosterone was reduced only in the males receiving the 2 highest doses of DIA. Serum estrone was increased in the 2 highest doses of the DACT group, while serum estradiol was not different in any group. No differences were observed in any of the thyroid measures. In summary, the metabolites of ATR delay puberty in a manner similar to that observed in the previous study testing atrazine. These data also suggest that the 3 chlorinated metabolites are similar to ATR, by affecting the CNS control of the pituitary/gonadal axis and subsequent development of the reproductive tract.


Asunto(s)
Atrazina/toxicidad , Herbicidas/toxicidad , Antagonistas de Hormonas/toxicidad , Maduración Sexual/efectos de los fármacos , Glándula Tiroides/efectos de los fármacos , Animales , Atrazina/análogos & derivados , Atrazina/metabolismo , Peso Corporal/efectos de los fármacos , Estradiol/sangre , Genitales Masculinos/efectos de los fármacos , Genitales Masculinos/patología , Herbicidas/metabolismo , Antagonistas de Hormonas/metabolismo , Masculino , Tamaño de los Órganos/efectos de los fármacos , Adenohipófisis/efectos de los fármacos , Adenohipófisis/patología , Ratas , Ratas Wistar , Maduración Sexual/fisiología , Testosterona/sangre , Glándula Tiroides/fisiopatología , Hormonas Tiroideas/sangre , Triazinas/toxicidad
17.
Toxicol Sci ; 76(1): 190-200, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12970575

RESUMEN

We showed previously that the chlorotriazine herbicide, atrazine (ATR), delays the onset of pubertal development in female rats. ATR and its biotransformation by-products are present in soil and groundwater. Since current maximum contaminant levels are set only for ATR, it is important to determine whether these by-products can also alter pubertal development and possibly pose a cumulative exposure hazard. We evaluated the effects of two ATR by-products, diamino-s-chlorotriazine (DACT) and hydroxyatrazine (OH-ATR), and a structurally similar chlorotriazine, propazine (PRO), on female pubertal development. Rats were gavaged from postnatal days (PNDs) 22 through PND 41 with DACT (16.7, 33.8, 67.5, 135 mg/kg), OH-ATR (22.8, 45.7, 91.5, 183 mg/kg), or PRO (13, 26.7, 53, 106.7, 213 mg/kg). The dose range for each chemical was selected as the molar equivalent of ATR (12.5, 25, 50, 100, 200 mg/kg). The females were monitored daily for vaginal opening (VO) and killed on PND 41. DACT, a by-product of ATR that occurs in the environment and is also the primary chlorinated metabolite of ATR in animal tissue, delayed VO by 3.2, 4.8, and 7.6 days compared to the controls (33.1 +/- 0.4 (SE) days of age) following exposure to 33.8, 67.5, and 135 mg/kg, respectively. The no effect level (NOEL) for DACT (16.7 mg/kg) was identical to the equimolar NOEL for ATR (25 mg/kg). Although the body weight (BW) on PND 41 was reduced by the high dose of DACT (8.4% reduction), this reduction did not exceed the criteria for selecting the maximum tolerated dose (e.g., a dose that causes >10% decrease in BW at necropsy). None of the lower doses of DACT caused a significant difference in BW gain. Additionally, 33.8, 67.5, and 135 mg/kg of DACT significantly increased the BW on the day of VO. PRO (107 or 213 mg/kg) delayed VO by 4 days but did not alter the BW on PND 41. While no significant delays in pubertal development were observed in two separate dose-response studies with doses ranging up to 183 mg/kg (OH-ATR), a minor but statistically significant delay in the onset of puberty in a pilot study using OH-ATR raises the possibility that an effect might occur following exposure to higher doses. However, it is clear from these data that OH-ATR has a much lower potency when compared with equimolar doses of DACT and PRO. Together, these data demonstrate that PRO and DACT can delay the onset of puberty in the female rat at doses equimolar to ATR and provide the scientific basis for the use of additivity in the upcoming risk assessments.


Asunto(s)
Atrazina/análogos & derivados , Atrazina/toxicidad , Herbicidas/metabolismo , Efectos Tardíos de la Exposición Prenatal , Maduración Sexual/efectos de los fármacos , Triazinas/toxicidad , Vagina/efectos de los fármacos , Administración Oral , Animales , Atrazina/metabolismo , Biotransformación , Peso Corporal/efectos de los fármacos , Femenino , Nivel sin Efectos Adversos Observados , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Ratas , Ratas Wistar , Hormonas Tiroideas/sangre , Triazinas/metabolismo , Vagina/crecimiento & desarrollo
18.
Toxicol Sci ; 78(1): 144-55, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14999130

RESUMEN

DE-71, a commercial mixture, was used to test the sensitivity of the female and male pubertal protocol to detect thyroid active chemicals. These protocols are being evaluated for the U.S. EPA's Endocrine Disruptor Screening Program as part of a Tier I Screening Battery. To examine the ability of these protocols to screen for chemicals that induce the clearance of thyroid hormone, we examined male and female Wistar rats following DE-71 exposure. Rats were gavaged daily with 0, 3, 30, or 60 mg/kg DE in corn oil from postnatal day (PND) 23-53 in the male or PND 22-41 in the female. The temporal effects of DE-71 on liver enzymes and thyroid hormones were measured in another group of males and females following only 5 days of dosing (PND 21 to 26 in females and PND 23 to 28 in males). Serum T4 was significantly decreased at 30 and 60 mg/kg following the 5-day exposures and in the 21-day exposed females. Doses of 3, 30, and 60 mg/kg decreased T4 in 31-day exposed males. Serum T3 was decreased and TSH elevated by 30 and 60 mg/kg in the 31-day exposed males only. Decreased colloid area and increased follicular cell heights (indicative of the hypothyroid state) were observed in thyroids of the 60 mg/kg groups of 20- and 31-day exposed female and males. Increased liver-to-body weight ratios coincided with a significant induction of uridinediphosphate-glucuronosyltransferase (UDGPT; two to four-fold), and ethoxy- and pentoxy-resorufin-O-deethylase (EROD and PROD) at the two highest doses in all exposures. Of the androgen dependent tissues in the 31-day exposed males, seminal vesicle (SV) and ventral prostate (VP) weights were reduced at 60 mg/kg, while testes and epididymal weights were not affected. Preputial separation (PPS) was also significantly delayed by doses of 30 and 60 mg/kg. In the female, the 60 mg/kg dose also caused a significant delay in the age of vaginal opening. Based upon the thyroid hormone response data, this study provides evidence that the 31-day alternative Tier 1 male protocol is a more sensitive test protocol than the 5-day or female pubertal protocol for thyrotoxic agents that act via up-regulation of hepatic metabolism. This apparent greater sensitivity may be due a greater body burden attained following the longer dosing regimen as compared with that of the female protocol, or to gender specific differences in thyroid hormone metabolism. Also, the delay in PPS and reduction in SV and VP weights may indicate a modification or inhibition of endogenous androgenic stimulation directly by DE-71 or a secondary effect that occurs in response to a DE-induced change in thyroid hormones.


Asunto(s)
Mezclas Complejas/toxicidad , Enfermedades del Sistema Endocrino/inducido químicamente , Hidrocarburos Bromados/toxicidad , Éteres Fenílicos/toxicidad , Bifenilos Polibrominados/toxicidad , Maduración Sexual/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Determinación de Punto Final , Femenino , Éteres Difenilos Halogenados , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Tamaño de los Órganos/efectos de los fármacos , Pene/efectos de los fármacos , Pene/crecimiento & desarrollo , Embarazo , Radioinmunoensayo , Ratas , Ratas Wistar , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/patología , Hormonas Tiroideas/sangre , Pruebas de Toxicidad , Vagina/efectos de los fármacos , Vagina/crecimiento & desarrollo
19.
J Toxicol Sci ; 27(5): 449-57, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12533915

RESUMEN

A significant negative correlation between testicular interstitial cell tumors and pituitary tumors in control male F344 rats has been reported associated with the number of animals per cage. Change in numbers of animals per cage may cause stress and increased serum corticosteroids that can impair testosterone synthesis by interstitial cells. Eventual atrophy of interstitial cells may result in pituitary hyperfunction and tumor development. For relevant risk assessments, understanding the effect husbandry has on cellular processes is necessary. Twenty-four 6-week-old male F344 rats were housed individually, as pairs, or as trios for 13 weeks. Measured parameters included feed consumption, body and organ weights, hemograms, hormonal levels, histopathology, and cellular kinetics in the pituitary and testicle. Several caging-related differences occurred, that, although not statistically different, could be biologically significant; these included increased serum levels of estradiol, progesterone, and corticosterone; increased spermatogonial proliferation; decreased apoptosis within seminiferous tubules; and increased BrdU immunoreactivity of the interstitial cells. The statistically significant decrease in lymphocyte numbers correlated with the associated increase in corticosterone levels. This study indicates that the number of animals in a cage is associated with hormonal and cellular kinetic changes in the pituitary and testes, which could influence the incidence of tumors in these organs.


Asunto(s)
Hormonas/fisiología , Vivienda para Animales , Pulmón/citología , Animales , Antimetabolitos , Peso Corporal/fisiología , Bromodesoxiuridina , División Celular/fisiología , Corticosterona/sangre , Ingestión de Alimentos/fisiología , Estradiol/sangre , Cinética , Masculino , Neoplasias Hipofisarias/epidemiología , Neoplasias Hipofisarias/patología , Progesterona/sangre , Ratas , Ratas Endogámicas F344 , Neoplasias Testiculares/epidemiología , Neoplasias Testiculares/patología
20.
Reprod Toxicol ; 36: 71-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23261820

RESUMEN

Triclosan (TCS), an antibacterial, has been shown to be an endocrine disruptor in the rat. We reported previously that TCS potentiated the estrogenic effect of ethinyl estradiol (EE) on uterine growth in rats exposed to EE and TCS in the uterotrophic assay, whereas TCS alone had no effect. To further characterize this potentiation, we evaluated the effect of co-exposure with lower doses of EE that are comparable to the concentrations in hormone replacement regimens and began to assess the mechanisms by which this potentiation occurs. Changes in uterine weight, epithelial cell growth, and estrogen-sensitive gene expression were assessed. TCS expectedly enhanced the uterotrophic response to EE, however at significantly lower doses of EE. Similarly, TCS increased the EE-induced stimulation of epithelial cell height following cotreatment. Cotreatment also enhanced the estrogen-induced change in gene expression, which was reversed with an ER antagonist. Furthermore, the TCS-induced potentiation was independent of ER activation, as no effects were observed in the ER TA assay.


Asunto(s)
Disruptores Endocrinos/toxicidad , Estrógenos/agonistas , Etinilestradiol/agonistas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Lesiones Precancerosas/inducido químicamente , Triclosán/toxicidad , Útero/efectos de los fármacos , Animales , Antibacterianos/administración & dosificación , Antibacterianos/antagonistas & inhibidores , Antibacterianos/toxicidad , Forma de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Disruptores Endocrinos/administración & dosificación , Disruptores Endocrinos/química , Antagonistas de Estrógenos/farmacología , Antagonistas de Estrógenos/uso terapéutico , Terapia de Reemplazo de Estrógeno/efectos adversos , Estrógenos/administración & dosificación , Estrógenos/efectos adversos , Estrógenos/farmacología , Etinilestradiol/efectos adversos , Etinilestradiol/antagonistas & inhibidores , Etinilestradiol/farmacología , Femenino , Tamaño de los Órganos/efectos de los fármacos , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Lesiones Precancerosas/prevención & control , Distribución Aleatoria , Ratas , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Triclosán/administración & dosificación , Triclosán/antagonistas & inhibidores , Útero/crecimiento & desarrollo , Útero/metabolismo , Útero/patología , Destete
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA