Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Am J Public Health ; 108(S3): S179-S182, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30192670

RESUMO

Following Hurricane Katrina, the uniformed US Public Health Service created an updated system through which its officers participated in emergency responses. The Rapid Deployment Force (RDF) concept, begun in 2006, involved five teams of officers with diverse clinical and public health skill sets organized into an incident command system led by a team commander. Each team can deploy within 12 hours, according to a defined but flexible schedule. The core RDF mission is to set up and provide care for up to 250 patients, primarily persons with chronic diseases or disabilities, in a temporary federal medical station. Between 2006 and 2016, the RDF 3 team deployed multiple times in response to natural disasters and public health emergencies. Notable responses included Hurricane Sandy in 2012, the unaccompanied children mission in 2014, and the Louisiana floods in 2016. Lessons learned from the RDF 3 experience include the need for both clinical and public health capacity, the value of having special mental health resources, the benefits of collaboration with other federal medical responders, and recognition of the large burden of chronic disease management issues following natural disasters.


Assuntos
Planejamento em Desastres , Desastres/história , Saúde Pública , Tempestades Ciclônicas/história , Inundações/história , História do Século XXI , Humanos , Louisiana , New Jersey
2.
Crit Rev Toxicol ; 44(7): 600-17, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25068490

RESUMO

Lipophilic persistent environmental chemicals (LPECs) have the potential to accumulate within a woman's body lipids over the course of many years prior to pregnancy, to partition into human milk, and to transfer to infants upon breastfeeding. As a result of this accumulation and partitioning, a breastfeeding infant's intake of these LPECs may be much greater than his/her mother's average daily exposure. Because the developmental period sets the stage for lifelong health, it is important to be able to accurately assess chemical exposures in early life. In many cases, current human health risk assessment methods do not account for differences between maternal and infant exposures to LPECs or for lifestage-specific effects of exposure to these chemicals. Because of their persistence and accumulation in body lipids and partitioning into breast milk, LPECs present unique challenges for each component of the human health risk assessment process, including hazard identification, dose-response assessment, and exposure assessment. Specific biological modeling approaches are available to support both dose-response and exposure assessment for lactational exposures to LPECs. Yet, lack of data limits the application of these approaches. The goal of this review is to outline the available approaches and to identify key issues that, if addressed, could improve efforts to apply these approaches to risk assessment of lactational exposure to these chemicals.


Assuntos
Poluentes Ambientais/análise , Exposição Materna , Leite Humano/química , Medição de Risco , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Modelos Teóricos , Método de Monte Carlo , Gravidez , Ratos , Projetos de Pesquisa
3.
Inhal Toxicol ; 26(10): 598-619, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25144475

RESUMO

Ethanol (EtOH) exposure induces a variety of concentration-dependent neurological and developmental effects in the rat. Physiologically-based pharmacokinetic (PBPK) models have been used to predict the inhalation exposure concentrations necessary to produce blood EtOH concentrations (BEC) in the range associated with these effects. Previous laboratory reports often lacked sufficient detail to adequately simulate reported exposure scenarios associated with BECs in this range, or lacked data on the time-course of EtOH in target tissues (e.g. brain, liver, eye, fetus). To address these data gaps, inhalation studies were performed at 5000, 10 000, and 21 000 ppm (6 h/d) in non-pregnant female Long-Evans (LE) rats and at 21 000 ppm (6.33 h/d) for 12 d of gestation in pregnant LE rats to evaluate our previously published PBPK models at toxicologically-relevant blood and tissue concentrations. Additionally, nose-only and whole-body plethysmography studies were conducted to refine model descriptions of respiration and uptake within the respiratory tract. The resulting time-course and plethysmography data from these in vivo studies were compared to simulations from our previously published models, after which the models were recalibrated to improve descriptions of tissue dosimetry by accounting for dose-dependencies in pharmacokinetic behavior. Simulations using the recalibrated models reproduced these data from non-pregnant, pregnant, and fetal rats to within a factor of 2 or better across datasets, resulting in a suite of model structures suitable for simulation of a broad range of EtOH exposure scenarios.


Assuntos
Etanol/farmacocinética , Exposição por Inalação , Exposição Materna , Troca Materno-Fetal/fisiologia , Modelos Biológicos , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Testes Respiratórios , Relação Dose-Resposta a Droga , Etanol/sangue , Etanol/toxicidade , Olho/embriologia , Olho/metabolismo , Feminino , Sangue Fetal/metabolismo , Idade Gestacional , Exposição por Inalação/efeitos adversos , Exposição por Inalação/análise , Cinética , Fígado/embriologia , Fígado/metabolismo , Exposição Materna/efeitos adversos , Troca Materno-Fetal/efeitos dos fármacos , Pletismografia , Gravidez , Ratos Long-Evans
4.
Risk Anal ; 34(2): 356-66, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23901895

RESUMO

Physiologically-based pharmacokinetic (PBPK) models are often submitted to or selected by agencies, such as the U.S. Environmental Protection Agency (U.S. EPA) and Agency for Toxic Substances and Disease Registry, for consideration for application in human health risk assessment (HHRA). Recently, U.S. EPA evaluated the human PBPK models for perchlorate and radioiodide for their ability to estimate the relative sensitivity of perchlorate inhibition on thyroidal radioiodide uptake for various population groups and lifestages. The most well-defined mode of action of the environmental contaminant, perchlorate, is competitive inhibition of thyroidal iodide uptake by the sodium-iodide symporter (NIS). In this analysis, a six-step framework for PBPK model evaluation was followed, and with a few modifications, the models were determined to be suitable for use in HHRA to evaluate relative sensitivity among human lifestages. Relative sensitivity to perchlorate was determined by comparing the PBPK model predicted percent inhibition of thyroidal radioactive iodide uptake (RAIU) by perchlorate for different lifestages. A limited sensitivity analysis indicated that model parameters describing urinary excretion of perchlorate and iodide were particularly important in prediction of RAIU inhibition; therefore, a range of biologically plausible values available in the peer-reviewed literature was evaluated. Using the updated PBPK models, the greatest sensitivity to RAIU inhibition was predicted to be the near-term fetus (gestation week 40) compared to the average adult and other lifestages; however, when exposure factors were taken into account, newborns were found to be populations that need further evaluation and consideration in a risk assessment for perchlorate.


Assuntos
Modelos Biológicos , Farmacocinética , Adulto , Fatores Etários , Criança , Feminino , Humanos , Recém-Nascido , Radioisótopos do Iodo/metabolismo , Percloratos/farmacocinética , Gravidez , Medição de Risco , Glândula Tireoide/metabolismo
5.
Toxicol Sci ; 194(2): 121-137, 2023 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-37228093

RESUMO

Per- and polyfluoroalkyl substances (PFAS) are a class of man-made chemicals that are persistent in the environment. They can be transferred across the placenta to fetuses and through human milk to infants. The American Academy of Pediatrics advises that the benefits of breastfeeding infants almost always outweigh the potential risks of harm from environmental chemicals. However, there are few chemical-specific summaries of the potential harms of exposure to PFAS during the neonatal period through breastfeeding. This systematic review explores whether exposure to PFAS through breastfeeding is associated with adverse health outcomes among infants and children using evidence from human and animal studies. Systematic searches identified 4297 unique records from 7 databases. The review included 37 total articles, including 9 animal studies and 1 human study measuring the direct contribution of exposure of the infant or pup through milk for any health outcome. Animal studies provided evidence of associations between exposure to PFOA through breastfeeding and reduced early life body weight gain, mammary gland development, and thyroid hormone levels. They also provided limited evidence of associations between PFOS exposure through breastfeeding with reduced early life body weight gain and cellular changes in the hippocampus. The direct relevance of any of these outcomes to human health is uncertain, and it is possible that many adverse health effects of exposure through breastfeeding have not yet been studied. This review documents the current state of science and highlights the need for future research to guide clinicians making recommendations on infant feeding.


Assuntos
Ácidos Alcanossulfônicos , Poluentes Ambientais , Fluorocarbonos , Lactente , Recém-Nascido , Gravidez , Feminino , Animais , Humanos , Criança , Aleitamento Materno , Saúde Pública , Leite Humano , Aumento de Peso , Fluorocarbonos/toxicidade , Poluentes Ambientais/toxicidade
6.
Inhal Toxicol ; 24(11): 698-722, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22954395

RESUMO

Biofuel blends of 10% ethanol (EtOH) and gasoline are common in the USA, and higher EtOH concentrations are being considered (15-85%). Currently, no physiologically-based pharmacokinetic (PBPK) models are available to describe the kinetics of EtOH-based biofuels. PBPK models were developed to describe life-stage differences in the kinetics of EtOH alone in adult, pregnant, and neonatal rats for inhalation, oral, and intravenous routes of exposure, using data available in the open literature. Whereas ample data exist from gavage and intravenous routes of exposure, kinetic data from inhalation exposures are limited, particularly at concentrations producing blood and target tissue concentrations associated with developmental neurotoxicity. Compared to available data, the three models reported in this paper accurately predicted the kinetics of EtOH, including the absorption, peak concentration, and clearance across multiple datasets. In general, model predictions for adult and pregnant animals matched inhalation and intravenous datasets better than gavage data. The adult model was initially better able to predict the time-course of blood concentrations than was the neonatal model. However, after accounting for age-related changes in gastric uptake using the calibrated neonate model, simulations consistently reproduced the early kinetic behavior in blood. This work provides comprehensive multi-route life-stage models of EtOH pharmacokinetics and represents a first step in development of models for use with gasoline-EtOH blends, with additional potential applicability in investigation of the pharmacokinetics of EtOH abuse, addiction, and toxicity.


Assuntos
Etanol/farmacocinética , Modelos Biológicos , Animais , Animais Recém-Nascidos , Biocombustíveis , Simulação por Computador , Vias de Administração de Medicamentos , Etanol/administração & dosagem , Etanol/metabolismo , Feminino , Gravidez , Ratos
7.
Environ Int ; 111: 260-267, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29325971

RESUMO

BACKGROUND: Multiple agencies have developed health-based toxicity values for exposure to perfluorooctanoic acid (PFOA). Although PFOA exposure occurs in utero and through breastfeeding, current health-based toxicity values have not been derived using fetal or child dosimetry. Therefore, current values may underestimate the potential risks to fetuses and nursing infants. OBJECTIVE: Using fetal and child dosimetry, we aimed to calculate PFOA maternal human equivalent doses (HEDs), corresponding to a developmental mouse study lowest observed adverse effect level (LOAEL, 1mg/kg/day). Further, we investigated the impact of breastfeeding duration and PFOA half-life on the estimated HEDs. METHODS: First, a pharmacokinetic model of pregnancy and lactation in mice was used to estimate plasma PFOA levels in pups following a maternal exposure to 1mg PFOA/kg/day for gestational days 1-17. Four plasma PFOA concentration metrics were estimated in pups: i) average prenatal; ii) average postnatal; iii) average overall (prenatal and postnatal); and iv) maximum. Then, Monte Carlo simulations were performed using a pharmacokinetic model of pregnancy and lactation in humans to generate distributions of maternal HEDs that would result in fetal/child plasma levels equivalent to those estimated in pups using the mouse model. Median (HED50) and 1st percentile (HED01) of calculated HEDs were calculated. RESULTS: Estimated PFOA maternal HED50s ranged from 3.0×10-4 to 1.1×10-3mg/kg/day and HED01s ranged from 4.7×10-5 to 2.1×10-4mg/kg/day. All calculated HEDs were lower than the HED based on adult dosimetry derived by the Environmental Protection Agency (EPA) (5.3×10-3mg/kg/day). CONCLUSION: Our results suggest that fetal/child dosimetry should be considered when deriving health-based toxicity values for potential developmental toxicants.


Assuntos
Aleitamento Materno , Caprilatos/farmacocinética , Fluorocarbonos/farmacocinética , Exposição Materna , Modelos Biológicos , Adulto , Animais , Caprilatos/toxicidade , Criança , Feminino , Feto , Fluorocarbonos/toxicidade , Meia-Vida , Humanos , Lactação , Camundongos , Gravidez
8.
Toxicol Sci ; 97(2): 308-17, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17379623

RESUMO

The objective of this research was to characterize the disturbances in the hypothalamic-pituitary-thyroid (HPT) axis resulting from exposure to a binary mixture, 3,3',4,4',5-pentachlorobiphenyl (PCB126) and perchlorate (ClO(4)(-)), known to cause hypothyroidism by different modes of action. Two studies were conducted to determine the HPT axis effects of ClO(4)(-) on adult male Sprague-Dawley rats pretreated with PCB126. In dosing study I, rats were administered a single oral dose of PCB126 (0, 7.5, or 75 microg/kg) on day 0 and 9 days later ClO(4)(-) (0, 0.01, 0.1, or 1 mg/kg day) was added to the drinking water until euthanasia on day 22. Significant dose-dependent trends were found for all thyroid function indices measured following ClO(4)(-) in drinking water for 14 days. Seventy-five micrograms PCB126/kg resulted in a significant increase in hepatic T(4)-glucuronide formation, causing a decline in serum thyroxine and fT(4), and resulting in increased serum thyroid-stimulating hormone (TSH). Serum TSH was also increased in animals that received 7.5 microg PCB126/kg; no other HPT axis alterations were found in these animals. When pretreated with PCB126, the ClO(4)(-) dose trends disappeared, suggesting a less than additive effect on the HPT axis. In dosing study II, animals were given lower doses of PCB126 (0, 0.075, 0.75, or 7.5 microg/kg) on day 0, and followed with ClO(4)(-) (0 or 0.01 mg/kg day) in drinking water beginning on day 1 and continuing for several days to explore transient HPT axis effects. No statistical effects were seen for PCB126 or ClO(4)(-) alone, and no perturbations were found when administered sequentially in dosing study II. In conclusion, these studies demonstrate that HPT axis disturbances following exposure to ClO(4)(-) are less than additive when pretreated with relatively high doses of PCB126. At relatively low doses, at or near the no-observed-effect-level for PCB126 and ClO(4)(-), no interactions between the chemicals occur.


Assuntos
Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Percloratos/toxicidade , Bifenilos Policlorados/toxicidade , Compostos de Amônio Quaternário/toxicidade , Glândula Tireoide/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Hormônios/sangue , Iodetos/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Simportadores/biossíntese , Simportadores/fisiologia , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Tireotropina/sangue
9.
PLoS One ; 11(3): e0149300, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26930410

RESUMO

The Institute of Medicine recommends that lactating women ingest 290 µg iodide/d and a nursing infant, less than two years of age, 110 µg/d. The World Health Organization, United Nations Children's Fund, and International Council for the Control of Iodine Deficiency Disorders recommend population maternal and infant urinary iodide concentrations ≥ 100 µg/L to ensure iodide sufficiency. For breast milk, researchers have proposed an iodide concentration range of 150-180 µg/L indicates iodide sufficiency for the mother and infant, however no national or international guidelines exist for breast milk iodine concentration. For the first time, a lactating woman and nursing infant biologically based model, from delivery to 90 days postpartum, was constructed to predict maternal and infant urinary iodide concentration, breast milk iodide concentration, the amount of iodide transferred in breast milk to the nursing infant each day and maternal and infant serum thyroid hormone kinetics. The maternal and infant models each consisted of three sub-models, iodide, thyroxine (T4), and triiodothyronine (T3). Using our model to simulate a maternal intake of 290 µg iodide/d, the average daily amount of iodide ingested by the nursing infant, after 4 days of life, gradually increased from 50 to 101 µg/day over 90 days postpartum. The predicted average lactating mother and infant urinary iodide concentrations were both in excess of 100 µg/L and the predicted average breast milk iodide concentration, 157 µg/L. The predicted serum thyroid hormones (T4, free T4 (fT4), and T3) in both the nursing infant and lactating mother were indicative of euthyroidism. The model was calibrated using serum thyroid hormone concentrations for lactating women from the United States and was successful in predicting serum T4 and fT4 levels (within a factor of two) for lactating women in other countries. T3 levels were adequately predicted. Infant serum thyroid hormone levels were adequately predicted for most data. For moderate iodide deficient conditions, where dietary iodide intake may range from 50 to 150 µg/d for the lactating mother, the model satisfactorily described the iodide measurements, although with some variation, in urine and breast milk. Predictions of serum thyroid hormones in moderately iodide deficient lactating women (50 µg/d) and nursing infants did not closely agree with mean reported serum thyroid hormone levels, however, predictions were usually within a factor of two. Excellent agreement between prediction and observation was obtained for a recent moderate iodide deficiency study in lactating women. Measurements included iodide levels in urine of infant and mother, iodide in breast milk, and serum thyroid hormone levels in infant and mother. A maternal iodide intake of 50 µg/d resulted in a predicted 29-32% reduction in serum T4 and fT4 in nursing infants, however the reduced serum levels of T4 and fT4 were within most of the published reference intervals for infant. This biologically based model is an important first step at integrating the rapid changes that occur in the thyroid system of the nursing newborn in order to predict adverse outcomes from exposure to thyroid acting chemicals, drugs, radioactive materials or iodine deficiency.


Assuntos
Aleitamento Materno , Suplementos Nutricionais/análise , Iodetos/análise , Iodetos/urina , Lactação , Leite Humano/química , Simulação por Computador , Feminino , Humanos , Recém-Nascido , Iodetos/administração & dosagem , Modelos Biológicos , Tiroxina/sangue , Tri-Iodotironina/sangue
11.
Toxicol Sci ; 143(2): 512-35, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25410581

RESUMO

To provide useful alternatives to in vivo animal studies, in vitro assays for dose-response assessments of xenobiotic chemicals must use concentrations in media and target tissues that are within biologically-plausible limits. Determining these concentrations is a complex matter, which can be facilitated by applying physiologically-based pharmacokinetic (PBPK) models in an in vitro to in vivo extrapolation (IVIVE) paradigm. We used ethanol (EtOH), a ubiquitous chemical with defined metrics for in vivo and in vitro embryotoxicity, as a model chemical to evaluate this paradigm. A published series of life-stage PBPK models for rats was extended to mice, yielding simulations that adequately predicted in vivo blood EtOH concentrations (BECs) from oral, intraperitoneal, and intravenous routes in nonpregnant and pregnant adult mice. The models were then extrapolated to nonpregnant and pregnant humans, replicating BEC data within a factor of two. The rodent models were then used to conduct IVIVEs for rodent and whole-embryo culture embryotoxicity data (neural tube closure defects, morphological changes). A second IVIVE was conducted for exposure scenarios in pregnant women during critical windows of susceptibility for developmental toxicity, such as the first 6-to-8 weeks (prerecognition period) or mid-to-late pregnancy period, when EtOH consumption is associated with fetal alcohol spectrum disorders. Incorporation of data from human embryonic stem cell studies led to a model-supported linkage of in vitro concentrations with plausible exposure ranges for pregnant women. This effort demonstrates benefits and challenges associated with use of multispecies PBPK models to estimate in vivo tissue concentrations associated with in vitro embryotoxicity studies.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Etanol/farmacocinética , Etanol/toxicidade , Desenvolvimento Fetal/efeitos dos fármacos , Exposição Materna , Modelos Biológicos , Simulação por Computador , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Etanol/administração & dosagem , Etanol/sangue , Feminino , Idade Gestacional , Humanos , Valor Preditivo dos Testes , Gravidez , Especificidade da Espécie , Distribuição Tecidual
12.
Toxicol Sci ; 132(1): 75-86, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23288054

RESUMO

A biologically based dose-response (BBDR) model for the hypothalamic-pituitary thyroid (HPT) axis in the lactating rat and nursing pup was developed to describe the perturbations caused by iodide deficiency on the HPT axis. Model calibrations, carried out by adjusting key model parameters, were used as a technique to evaluate HPT axis adaptations to dietary iodide intake in euthyroid (4.1-39 µg iodide/day) and iodide-deficient (0.31 and 1.2 µg iodide/day) conditions. Iodide-deficient conditions in both the dam and the pup were described with increased blood flow to the thyroid gland, TSH-mediated increase in thyroidal uptake of iodide and binding of iodide in the thyroid gland (organification), and, in general, reduced thyroid hormone production and metabolism. Alterations in thyroxine (T4) homeostasis were more apparent than for triiodothyronine (T3). Model-predicted average daily area-under-the-serum-concentration-curve (nM-day) values for T4 at steady state in the dam and pup decreased by 14-15% for the 1.2 µg iodide/day iodide-deficient diet and 42-52% for the 0.31 µg iodide/day iodide-deficient diet. In rat pups that were iodide deficient during gestation and lactation, these decreases in serum T4 levels were associated with declines in thyroid hormone in the fetal brain and a suppression of synaptic responses in the hippocampal region of the brain of the adult offspring (Gilbert et al. , 2013).


Assuntos
Deficiências Nutricionais/fisiopatologia , Iodetos/metabolismo , Lactação , Modelos Biológicos , Animais , Calibragem , Relação Dose-Resposta a Droga , Sistema Hipotálamo-Hipofisário , Ratos , Glândula Tireoide/fisiopatologia , Hormônios Tireóideos/sangue
13.
Toxicol Sci ; 126(1): 5-15, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22045031

RESUMO

A panel of experts in physiologically based pharmacokinetic (PBPK) modeling and relevant quantitative methods was convened to describe and discuss model evaluation criteria, issues, and choices that arise in model application and computational tools for improving model quality for use in human health risk assessments (HHRAs). Although publication of a PBPK model in a peer-reviewed journal is a mark of good science, subsequent evaluation of published models and the supporting computer code is necessary for their consideration for use in HHRAs. Standardized model evaluation criteria and a thorough and efficient review process can reduce the number of review and revision iterations and hence the time needed to prepare a model for application. Efficient and consistent review also allows for rapid identification of needed model modifications to address HHRA-specific issues. This manuscript reports on the workshop where a process and criteria that were created for PBPK model review were discussed along with other issues related to model review and application in HHRA. Other issues include (1) model code availability, portability, and validity; (2) probabilistic (e.g., population-based) PBPK models and critical choices in parameter values to fully characterize population variability; and (3) approaches to integrating PBPK model outputs with other HHRA tools, including benchmark dose modeling. Two specific case study examples are provided to illustrate challenges that were encountered during the review and application process. By considering the frequent challenges encountered in the review and application of PBPK models during the model development phase, scientists may be better able to prepare their models for use in HHRAs.


Assuntos
Modelos Biológicos , Publicações Periódicas como Assunto , Farmacocinética , Medição de Risco/métodos , Animais , Humanos , Método de Monte Carlo , Farmacologia Clínica/métodos , Toxicologia/métodos
14.
Environ Health Perspect ; 117(5): 731-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19479014

RESUMO

BACKGROUND: Perchlorate (ClO4(-)) is an environmental contaminant known to disrupt the thyroid axis of many terrestrial and aquatic species. ClO4(-) competitively inhibits iodide uptake into the thyroid at the sodium/iodide symporter and disrupts hypothalamic-pituitary-thyroid (HPT) axis homeostasis in rodents. OBJECTIVE: We evaluated the proposed mode of action for ClO4(-)-induced rat HPT axis perturbations using a biologically based dose-response (BBDR) model of the HPT axis coupled with a physiologically based pharmacokinetic model of ClO4(-). METHODS: We configured a BBDR-HPT/ClO4(-) model to describe competitive inhibition of thyroidal uptake of dietary iodide by ClO4(-) and used it to simulate published adult rat drinking water studies. We compared model-predicted serum thyroid-stimulating hormone (TSH) and total thyroxine (TT4) concentrations with experimental observations reported in these ClO4(-) drinking water studies. RESULTS: The BBDR-HPT/ClO4(-) model failed to predict the ClO4(-)-induced onset of disturbances in the HPT axis. Using ClO4(-) inhibition of dietary iodide uptake into the thyroid, the model underpredicted both the rapid decrease in serum TT4 concentrations and the rise in serum TSH concentrations. CONCLUSIONS: Assuming only competitive inhibition of thyroidal uptake of dietary iodide, BBDR-HPT/ClO4(-) model calculations were inconsistent with the rapid decrease in serum TT4 and the corresponding increase in serum TSH. Availability of bound iodide in the thyroid gland governed the rate of hormone secretion from the thyroid. ClO4(-) is translocated into the thyroid gland, where it may act directly or indirectly on thyroid hormone synthesis/secretion in the rat. The rate of decline in serum TT4 in these studies after 1 day of treatment with ClO4(-) appeared consistent with a reduction in thyroid hormone production/secretion. This research demonstrates the utility of a biologically based model to evaluate a proposed mode of action for ClO4(-) in a complex biological process.


Assuntos
Transporte Biológico/efeitos dos fármacos , Iodetos/metabolismo , Percloratos/toxicidade , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Tireotropina/sangue , Tiroxina/sangue , Poluentes Químicos da Água/toxicidade , Animais , Masculino , Ratos
15.
Toxicol Sci ; 102(2): 241-53, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18178547

RESUMO

A biologically based dose-response (BBDR) model was developed for dietary iodide and the hypothalamic-pituitary-thyroid (HPT) axis in adult rats. This BBDR-HPT axis model includes submodels for dietary iodide, thyroid-stimulating hormone (TSH), and the thyroid hormones, T(4) and T(3). The submodels are linked together via key biological processes, including (1) the influence of T(4) on TSH production (the HPT axis negative feedback loop), (2) stimulation of thyroidal T(4) and T(3) production by TSH, (3) TSH upregulation of the thyroid sodium (Na(+))/iodide symporter, and (4) recycling of iodide from metabolism of thyroid hormones. The BBDR-HPT axis model was calibrated to predict steady-state concentrations of iodide, T(4), T(3), and TSH for the euthyroid rat whose dietary intake of iodide was 20 mug/day. Then the BBDR-HPT axis model was used to predict perturbations in the HPT axis caused by insufficient dietary iodide intake, and simulation results were compared to experimental findings. The BBDR-HPT axis model was successful in simulating perturbations in serum T(4), TSH, and thyroid iodide stores for low-iodide diets of 0.33-1.14 mug/day. Model predictions of serum T(3) concentrations were inconsistent with observations in some cases. BBDR-HPT axis model simulations show a steep dose-response relationship between dietary intake of iodide and serum T(4) and TSH when dietary iodide intake becomes insufficient (less than 2 mug/day) to sustain the HPT axis. This BBDR-HPT axis model can be linked with physiologically based pharmacokinetic models for thyroid-active chemicals to evaluate and predict dose-dependent HPT axis alterations based on hypothesized modes of action. To support continued development of this model, future studies should include time course data after perturbation of the HPT axis to capture changes in endogenous iodide, serum TSH, T(4), and T(3).


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Iodetos/farmacocinética , Iodo/deficiência , Glândula Tireoide/metabolismo , Animais , Dieta , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Iodetos/toxicidade , Iodo/metabolismo , Radioisótopos do Iodo , Modelos Biológicos , Ratos , Ratos Sprague-Dawley , Glândula Tireoide/efeitos dos fármacos , Tireotropina/metabolismo , Tiroxina/metabolismo , Tri-Iodotironina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA