1
|
Gonnabathula P, Choi MK, Li M, Kabadi SV, Fairman K. Utility of life stage-specific chemical risk assessments based on New Approach Methodologies (NAMs). Food Chem Toxicol 2024; 190:114789. [PMID: 38844066 DOI: 10.1016/j.fct.2024.114789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 06/17/2024]
Abstract
The safety assessments for chemicals targeted for use or expected to be exposed to specific life stages, including infancy, childhood, pregnancy and lactation, and geriatrics, need to account for extrapolation of data from healthy adults to these populations to assess their human health risk. However, often adequate and relevant toxicity or pharmacokinetic (PK) data of chemicals in specific life stages are not available. For such chemicals, New Approach Methodologies (NAMs), such as physiologically based pharmacokinetic (PBPK) modeling, biologically based dose response (BBDR) modeling, in vitro to in vivo extrapolation (IVIVE), etc. can be used to understand the variability of exposure and effects of chemicals in specific life stages and assess their associated risk. A life stage specific PBPK model incorporates the physiological and biochemical changes associated with each life stage and simulates their impact on the absorption, distribution, metabolism, and elimination (ADME) of these chemicals. In our review, we summarize the parameterization of life stage models based on New Approach Methodologies (NAMs) and discuss case studies that highlight the utility of a life stage based PBPK modeling for risk assessment. In addition, we discuss the utility of artificial intelligence (AI)/machine learning (ML) and other computational models, such as those based on in vitro data, as tools for estimation of relevant physiological or physicochemical parameters and selection of model. We also discuss existing gaps in the available toxicological datasets and current challenges that need to be overcome to expand the utility of NAMs for life stage-specific chemical risk assessment.
Collapse
Affiliation(s)
- Pavani Gonnabathula
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), US Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Me-Kyoung Choi
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), US Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Miao Li
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), US Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Shruti V Kabadi
- Center for Food Safety and Applied Nutrition (CFSAN), US Food and Drug Administration (FDA), College Park, MD, 20740, USA
| | - Kiara Fairman
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), US Food and Drug Administration (FDA), Jefferson, AR, 72079, USA.
| |
Collapse
|
2
|
Fisher J, Housand C, Mattie D, Nong A, Moreau M, Gilbert M. Towards translating in vitro measures of thyroid hormone system disruption to in vivo responses in the pregnant rat via a biologically based dose response (BBDR) model. Toxicol Appl Pharmacol 2023; 479:116733. [PMID: 37866708 DOI: 10.1016/j.taap.2023.116733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Despite the number of in vitro assays that have been recently developed to identify chemicals that interfere with the hypothalamic-pituitary-thyroid axis (HPT), the translation of those in vitro results into in vivo responses (in vitro to in vivo extrapolation, IVIVE) has received limited attention from the modeling community. To help advance this field a steady state biologically based dose response (BBDR) model for the HPT axis was constructed for the pregnant rat on gestation day (GD) 20. The BBDR HPT axis model predicts plasma levels of thyroid stimulating hormone (TSH) and the thyroid hormones, thyroxine (T4) and triiodothyronine (T3). Thyroid hormones are important for normal growth and development of the fetus. Perchlorate, a potent inhibitor of thyroidal uptake of iodide by the sodium iodide symporter (NIS) protein, was used as a case study for the BBDR HPT axis model. The inhibitory blocking of the NIS by perchlorate was associated with dose-dependent steady state decreases in thyroid hormone production in the thyroid gland. The BBDR HPT axis model predictions for TSH, T3, and T4 plasma concentrations in pregnant Sprague Dawley (SD) rats were within 2-fold of observations for drinking water perchlorate exposures ranging from 10 to 30,000 μg/kg/d. In Long Evans (LE) pregnant rats, for both control and perchlorate drinking water exposures, ranging from 85 to 82,000 μg/kg/d, plasma thyroid hormone and TSH concentrations were predicted within 2 to 3.4- fold of observations. This BBDR HPT axis model provides a successful IVIVE template for thyroid hormone disruption in pregnant rats.
Collapse
Affiliation(s)
| | - Conrad Housand
- Magnolia Sciences, Winter Springs, FL, United States of America
| | - David Mattie
- AFRL/711 HPW/RHBAF, WPAFB, OH, United States of America
| | - Andy Nong
- ScitoVation LLC, RTP, NC, United States of America
| | | | - Mary Gilbert
- Office of Research and Development, Center for Public Health and Environmental Assessment, US EPA, RTP, NC, United States of America
| |
Collapse
|
3
|
Haselman JT, Nichols JW, Mattingly KZ, Hornung MW, Degitz SJ. A biologically based computational model for the hypothalamic-pituitary-thyroid (HPT) axis in Xenopus laevis larvae. Math Biosci 2023; 362:109021. [PMID: 37201649 PMCID: PMC11556306 DOI: 10.1016/j.mbs.2023.109021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/28/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023]
Abstract
A biologically based computational model was developed to describe the hypothalamic-pituitary-thyroid (HPT) axis in developing Xenopus laevis larvae. The goal of this effort was to develop a tool that can be used to better understand mechanisms of thyroid hormone-mediated metamorphosis in X. laevis and predict organismal outcomes when those mechanisms are perturbed by chemical toxicants. In this report, we describe efforts to simulate the normal biology of control organisms. The structure of the model borrows from established models of HPT axis function in mammals. Additional features specific to X. laevis account for the effects of organism growth, growth of the thyroid gland, and developmental changes in regulation of thyroid stimulating hormone (TSH) by circulating thyroid hormones (THs). Calibration was achieved by simulating observed changes in stored and circulating levels of THs during a critical developmental window (Nieuwkoop and Faber stages 54-57) that encompasses widely used in vivo chemical testing protocols. The resulting model predicts that multiple homeostatic processes, operating in concert, can act to preserve circulating levels of THs despite profound impairments in TH synthesis. Represented in the model are several biochemical processes for which there are high-throughput in vitro chemical screening assays. By linking the HPT axis model to a toxicokinetic model of chemical uptake and distribution, it may be possible to use this in vitro effects information to predict chemical effects in X. laevis larvae resulting from defined chemical exposures.
Collapse
Affiliation(s)
- Jonathan T Haselman
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, 6201 Congdon Boulevard, Duluth, MN, 55804, United States of America.
| | - John W Nichols
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, 6201 Congdon Boulevard, Duluth, MN, 55804, United States of America
| | - Kali Z Mattingly
- SpecPro Professional Services (SPS), Contractor to U.S. Environmental Protection Agency, Great Lakes Toxicology and Ecology Division, 6201 Congdon Boulevard, Duluth, MN, 55804, United States of America
| | - Michael W Hornung
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, 6201 Congdon Boulevard, Duluth, MN, 55804, United States of America
| | - Sigmund J Degitz
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, 6201 Congdon Boulevard, Duluth, MN, 55804, United States of America
| |
Collapse
|
4
|
Bagga AD, Johnson BP, Zhang Q. A minimal human physiologically based kinetic model of thyroid hormones and chemical disruption of plasma thyroid hormone binding proteins. Front Endocrinol (Lausanne) 2023; 14:1168663. [PMID: 37305053 PMCID: PMC10248451 DOI: 10.3389/fendo.2023.1168663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/11/2023] [Indexed: 06/13/2023] Open
Abstract
The thyroid hormones (THs), thyroxine (T4) and triiodothyronine (T3), are under homeostatic control by the hypothalamic-pituitary-thyroid axis and plasma TH binding proteins (THBPs), including thyroxine-binding globulin (TBG), transthyretin (TTR), and albumin (ALB). THBPs buffer free THs against transient perturbations and distribute THs to tissues. TH binding to THBPs can be perturbed by structurally similar endocrine-disrupting chemicals (EDCs), yet their impact on circulating THs and health risks are unclear. In the present study, we constructed a human physiologically based kinetic (PBK) model of THs and explored the potential effects of THBP-binding EDCs. The model describes the production, distribution, and metabolism of T4 and T3 in the Body Blood, Thyroid, Liver, and Rest-of-Body (RB) compartments, with explicit consideration of the reversible binding between plasma THs and THBPs. Rigorously parameterized based on literature data, the model recapitulates key quantitative TH kinetic characteristics, including free, THBP-bound, and total T4 and T3 concentrations, TH productions, distributions, metabolisms, clearance, and half-lives. Moreover, the model produces several novel findings. (1) The blood-tissue TH exchanges are fast and nearly at equilibrium especially for T4, providing intrinsic robustness against local metabolic perturbations. (2) Tissue influx is limiting for transient tissue uptake of THs when THBPs are present. (3) Continuous exposure to THBP-binding EDCs does not alter the steady-state levels of THs, while intermittent daily exposure to rapidly metabolized TBG-binding EDCs can cause much greater disruptions to plasma and tissue THs. In summary, the PBK model provides novel insights into TH kinetics and the homeostatic roles of THBPs against thyroid disrupting chemicals.
Collapse
Affiliation(s)
- Anish D. Bagga
- Emory College of Arts and Sciences, Emory University, Atlanta, GA, United States
| | - Brian P. Johnson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, GA, Atlanta, United States
| |
Collapse
|
5
|
Fedoruk RS, Tesarivska UI, Kovalchuk II, Iskra RJ, Tsap M, Khrabko MI, Koleshchuk OI. The indices of thyroid system and metabolism of rats under the influence of nanocomposition based on iodine and citrate. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.03.092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
6
|
Wu H, Zhang W, Zhang Y, Kang Z, Miao X, Na X. Novel insights into di‑(2‑ethylhexyl)phthalate activation: Implications for the hypothalamus‑pituitary‑thyroid axis. Mol Med Rep 2021; 23:290. [PMID: 33649816 PMCID: PMC7930932 DOI: 10.3892/mmr.2021.11930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 09/09/2020] [Indexed: 11/06/2022] Open
Abstract
Di (2‑ethylhexyl) phthalate (DEHP), an environmental pollutant, is widely used as a plasticizer and causes serious pollution in the ecological environment. As previously reported, exposure to DEHP may cause thyroid dysfunction of the hypothalamic‑pituitary‑thyroid (HPT) axis. However, the underlying role of DEHP remains to be elucidated. The present study performed intragastrical administration of DEHP (150, 300 and 600 mg/kg) once a day for 90 consecutive days. DEHP‑stimulated oxidative stress increased the thyroid follicular cavity diameter and caused thyrocyte oedema. Furthermore, DEHP exposure altered mRNA and protein levels. Thus, DEHP may perturb TH homeostasis by affecting biosynthesis, biotransformation, bio‑transportation, receptor levels and metabolism through disruption of the HPT axis and activation of the thyroid‑stimulating hormone (TSH)/TSH receptor signaling pathway. These results identified the formerly unappreciated endocrine‑disrupting activities of phthalates and the molecular mechanisms of DEHP‑induced thyrotoxicity.
Collapse
Affiliation(s)
- Haoyu Wu
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Wanying Zhang
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
- Department of Logistics Support, Chengdu Blood Center, Chengdu, Sichuan 610041, P.R. China
| | - Yunbo Zhang
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zhen Kang
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
- Department of Environmental Hygiene, Harbin Center for Disease Control and Prevention, Harbin, Heilongjiang 150001, P.R. China
| | - Xinxiunan Miao
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaolin Na
- Department of Environmental Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
7
|
Volarath P, Zang Y, Kabadi SV. Application of Computational Methods for the Safety Assessment of Food Ingredients. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/978-3-030-16443-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
8
|
An evaluation of the USEPA Proposed Approaches for applying a biologically based dose-response model in a risk assessment for perchlorate in drinking water. Regul Toxicol Pharmacol 2019; 103:237-252. [DOI: 10.1016/j.yrtph.2019.01.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/18/2019] [Accepted: 01/20/2019] [Indexed: 12/18/2022]
|
9
|
Willemin ME, Lumen A. Characterization of the modes of action and dose-response relationship for thiocyanate on the thyroid hormone levels in rats using a computational approach. Toxicol Appl Pharmacol 2019; 365:84-100. [DOI: 10.1016/j.taap.2019.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 01/16/2023]
|
10
|
Repeated KI Prophylaxis in Case of Prolonged Exposure to Iodine Radioisotopes: Pharmacokinetic Studies in Adult Rats. Pharm Res 2018; 35:227. [PMID: 30298383 DOI: 10.1007/s11095-018-2515-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE To propose a new and effective dose regimen for stable potassium iodide (KI) repeated prophylaxis in case of prolonged exposure to radioactive iodine. METHODS The pharmacokinetics of iodine was determined in rats by compartmental analyses after intravenous and oral administrations of the optimal dose of 1 mg/kg KI, which was previously selected in a dose-effect study. The thyroid protection against iodine-125 incorporation was followed during 24 h after a single oral dosing of KI. A repeated KI prophylaxis was modeled using initial estimates of iodine pharmacokinetic parameters. RESULTS A dose regimen consisting in administrations of 1 mg/kg daily for 8 days was selected and studied. Plasma iodine concentrations predicted by simulation were verified by experimental data and varied after the third dose of KI between 174 and 1190 μg/l. The inhibition study of iodine-125 binding in the thyroid as a function of the time showed that the protection effect of KI could be correlated to stable iodine plasma concentrations. Hence, a theoretical decrease in iodine-125 thyroid uptake from 63 to 88% could be achieved in a 24 h-interval between two KI doses. CONCLUSION Given the satisfactory levels of thyroid protection, this dose regimen could be envisaged in order to extent KI indications for repeated prophylaxis.
Collapse
|
11
|
Dietrich JW, Midgley JEM, Hoermann R. Editorial: "Homeostasis and Allostasis of Thyroid Function". Front Endocrinol (Lausanne) 2018; 9:287. [PMID: 29922229 PMCID: PMC5996081 DOI: 10.3389/fendo.2018.00287] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- Johannes W. Dietrich
- Medical Department 1, Endocrinology and Diabetology, Bergmannsheil University Hospitals, Ruhr University of Bochum, Bochum, North Rhine-Westphalia, Germany
- Ruhr Centre of Rare Diseases (CeSER), Ruhr University of Bochum, Bochum, North Rhine-Westphalia, Germany
- Ruhr Centre of Rare Diseases (CeSER), Witten/Herdecke University, Bochum, North Rhine-Westphalia, Germany
- *Correspondence: Johannes W. Dietrich,
| | | | - Rudolf Hoermann
- Private Consultancy, Research and Development, Yandina, QLD, Australia
| |
Collapse
|
12
|
Phan G, Rebière F, Suhard D, Legrand A, Carpentier F, Sontag T, Souidi M, Jourdain JR, Agarande M, Renaud-Salis V. Optimal KI Prophylactic Dose Determination for Thyroid Radiation Protection After a Single Administration in Adult Rats. Dose Response 2017; 15:1559325817746558. [PMID: 29276472 PMCID: PMC5734494 DOI: 10.1177/1559325817746558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 12/05/2022] Open
Abstract
A dose–response study was performed in adult rats to select an optimal stable potassium iodide (KI) dose which could be implemented in repeated prophylaxis, in case of prolonged exposure to radioactive iodine. Increasing doses of KI were given orally to rats 1 hour before internal exposure simulated by I-125 injection. I-125 incorporation in the thyroid was measured by γ-spectrometry, and KI protection effect was modeled by pharmacological functions. The measurement method by inductively coupled plasma mass spectrometry previously developed for the quantification of stable iodine in urine was adapted to correlate KI effect with its distribution in the thyroid. More than 75% blockade of iodine I-125 incorporation in the thyroid was achieved for KI single doses above 0.5 to 0.7 mg/kg. Stable iodine content in the thyroid 24 hours after KI administration displayed a biphasic response, with a maximum level for a dose around 1 mg/kg. Besides, the urinary excretion of stable iodine is described by a sigmoid function. The change in the rate of iodine excretion for doses above 1 mg/kg KI suggests a body overload in iodine and corroborates a possible saturation of the thyroid. The results show that 1 mg/kg KI could be regarded as an optimal dose for thyroid protection.
Collapse
Affiliation(s)
- Guillaume Phan
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - François Rebière
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - David Suhard
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - Alexandre Legrand
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - Floriane Carpentier
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - Thibaud Sontag
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - Maâmar Souidi
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - Jean-René Jourdain
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - Michelle Agarande
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| | - Valérie Renaud-Salis
- Health Division, Institute for Radiation Protection and Nuclear Safety, Paris, France
| |
Collapse
|
13
|
Lin Z, Jaberi-Douraki M, He C, Jin S, Yang RSH, Fisher JW, Riviere JE. Performance Assessment and Translation of Physiologically Based Pharmacokinetic Models From acslX to Berkeley Madonna, MATLAB, and R Language: Oxytetracycline and Gold Nanoparticles As Case Examples. Toxicol Sci 2017; 158:23-35. [DOI: 10.1093/toxsci/kfx070] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
14
|
Willemin ME, Lumen A. Thiocyanate: a review and evaluation of the kinetics and the modes of action for thyroid hormone perturbations. Crit Rev Toxicol 2017. [DOI: 10.1080/10408444.2017.1281590] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Marie-Emilie Willemin
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Annie Lumen
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
15
|
Alayoubi A, Sullivan RD, Lou H, Patel H, Mandrell T, Helms R, Almoazen H. In Vivo Evaluation of Transdermal Iodide Microemulsion for Treating Iodine Deficiency Using Sprague Dawley Rats. AAPS PharmSciTech 2016; 17:618-30. [PMID: 26288943 DOI: 10.1208/s12249-015-0392-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/09/2015] [Indexed: 11/30/2022] Open
Abstract
The objective of this study was to evaluate the transdermal efficiency of iodide microemulsion in treating iodine deficiency using rats as an animal model. Animals were fed either iodine-deficient diet (20 μg/kg iodide) or control diet (200 μg/kg iodide) over a 17-month period. At month 14, iodide microemulsion was applied topically in iodine-deficient group and physiological evaluations of thyroid gland functions were characterized by monitoring the thyroid hormones (T3, T4), thyroid-stimulating hormone (TSH), iodide ion excretion in urine, and the overall rat body weights in both groups. Moreover, morphological evaluations of thyroid gland before and after treatment were performed by ultrasound imaging and through histological assessment. Prior to microemulsion treatment, the levels of T3, T4, and TSH in iodine-deficient group were statistically significant as compared to that in the control group. The levels of T3 and T4 increased while TSH level decreased significantly in iodine-deficient group within the first 4 weeks of treatment. After treatment, iodide concentration in urine increased significantly. There was no statistical difference in weight between the two groups. Ultrasound imaging and histological evaluations showed evidence of hyperplasia in iodine-deficient group. Topical iodide microemulsion has shown a promising potential as a novel delivery system to treat iodine deficiency.
Collapse
|
16
|
Fisher W, Wang J, George NI, Gearhart JM, McLanahan ED. Dietary Iodine Sufficiency and Moderate Insufficiency in the Lactating Mother and Nursing Infant: A Computational Perspective. PLoS One 2016; 11:e0149300. [PMID: 26930410 PMCID: PMC4773173 DOI: 10.1371/journal.pone.0149300] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/29/2016] [Indexed: 12/27/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- W. Fisher
- US FDA, National Center for Toxicological Research, 3900 NCTR Rd, Jefferson, Arkansas, 72079, United States of America
- * E-mail:
| | - Jian Wang
- US FDA, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Silver Springs, Maryland, 20993, United States of America
| | - Nysia I. George
- US FDA, National Center for Toxicological Research, 3900 NCTR Rd, Jefferson, Arkansas, 72079, United States of America
| | - Jeffery M. Gearhart
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, 2729 R Street, Bldg 837, Wright-Patterson AFB, Ohio, 43433, United States of America
- Wright State University Boonshoft School of Medicine, Dayton, Ohio, 45435, United States of America
| | - Eva D. McLanahan
- CDC/ATSDR, Division of Community Health Investigations, 4770 Buford HWY NE, Atlanta, Georgia, 30341, United States of America
| |
Collapse
|
17
|
Truter JC, van Wyk JH, Oberholster PJ, Botha AM, Luus-Powell WJ. The expression of selected genes linked to metabolic homeostasis in obese pansteatitis-suffering Mozambique tilapia, Oreochromis mossambicus (Peters). JOURNAL OF FISH DISEASES 2016; 39:69-85. [PMID: 25413848 DOI: 10.1111/jfd.12324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 10/06/2014] [Accepted: 10/06/2014] [Indexed: 06/04/2023]
Abstract
The Oreochromis mossambicus (Peters) population inhabiting Lake Loskop, South Africa, is characterized by a high incidence of obesity and pansteatitis. We investigated potential links between the impaired health of Lake Loskop O. mossambicus and the endocrine system by assessing the expression of selected genes associated with the thyroid and adrenal endocrine axes as well as peroxisome proliferator-activated receptor gamma (pparg). Moreover, contaminant-induced thyroid and/or metabolic modulation in Lake Loskop water was evaluated using juvenile O. mossambicus in laboratory exposures. The expression of thyroid hormone receptor alpha (thra) and type 2 deiodinase (dio2) was higher in Lake Loskop O. mossambicus than fish from another population, suggesting a degree of thyroid disruption. The altered gene expression may be a consequence, rather than cause of obesity. Expression of dio2 and pparg was higher in juvenile O. mossambicus exposed to unfiltered compared to filtered lake water, and our data suggest fasting as causative factor. Micro-organism abundance can therefore be a confounding factor in studies applying molecular markers to test for thyroid modulation by environmental waters. Pansteatitis was not a significant source of variance in the expression of any of the genes investigated, suggesting that the disease is not associated with disrupted endocrine signalling.
Collapse
Affiliation(s)
- J C Truter
- Department of Botany and Zoology, Stellenbosch University, Stellenbosch, South Africa
| | - J H van Wyk
- Department of Botany and Zoology, Stellenbosch University, Stellenbosch, South Africa
| | - P J Oberholster
- CSIR Natural Resources and the Environment, Stellenbosch, South Africa
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| | - A-M Botha
- Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - W J Luus-Powell
- Department of Biodiversity, University of Limpopo, Sovenga, South Africa
| |
Collapse
|
18
|
Axthammer QJ, Klapötke TM, Krumm B. Efficient Synthesis of Primary Nitrocarbamates of Sugar Alcohols: From Food to Energetic Materials. Chem Asian J 2015; 11:568-75. [DOI: 10.1002/asia.201501241] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Quirin J. Axthammer
- Energetic Materials Research Department of Chemistry Ludwig-Maximilians University of Munich Butenandtstr. 5–13 (D) 81377 Munich Germany
| | - Thomas M. Klapötke
- Energetic Materials Research Department of Chemistry Ludwig-Maximilians University of Munich Butenandtstr. 5–13 (D) 81377 Munich Germany
| | - Burkhard Krumm
- Energetic Materials Research Department of Chemistry Ludwig-Maximilians University of Munich Butenandtstr. 5–13 (D) 81377 Munich Germany
| |
Collapse
|
19
|
Lumen A, McNally K, George N, Fisher JW, Loizou GD. Quantitative global sensitivity analysis of a biologically based dose-response pregnancy model for the thyroid endocrine system. Front Pharmacol 2015; 6:107. [PMID: 26074819 PMCID: PMC4444753 DOI: 10.3389/fphar.2015.00107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/04/2015] [Indexed: 12/15/2022] Open
Abstract
A deterministic biologically based dose-response model for the thyroidal system in a near-term pregnant woman and the fetus was recently developed to evaluate quantitatively thyroid hormone perturbations. The current work focuses on conducting a quantitative global sensitivity analysis on this complex model to identify and characterize the sources and contributions of uncertainties in the predicted model output. The workflow and methodologies suitable for computationally expensive models, such as the Morris screening method and Gaussian Emulation processes, were used for the implementation of the global sensitivity analysis. Sensitivity indices, such as main, total and interaction effects, were computed for a screened set of the total thyroidal system descriptive model input parameters. Furthermore, a narrower sub-set of the most influential parameters affecting the model output of maternal thyroid hormone levels were identified in addition to the characterization of their overall and pair-wise parameter interaction quotients. The characteristic trends of influence in model output for each of these individual model input parameters over their plausible ranges were elucidated using Gaussian Emulation processes. Through global sensitivity analysis we have gained a better understanding of the model behavior and performance beyond the domains of observation by the simultaneous variation in model inputs over their range of plausible uncertainties. The sensitivity analysis helped identify parameters that determine the driving mechanisms of the maternal and fetal iodide kinetics, thyroid function and their interactions, and contributed to an improved understanding of the system modeled. We have thus demonstrated the use and application of global sensitivity analysis for a biologically based dose-response model for sensitive life-stages such as pregnancy that provides richer information on the model and the thyroidal system modeled compared to local sensitivity analysis.
Collapse
Affiliation(s)
- Annie Lumen
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration Jefferson, AR, USA
| | | | - Nysia George
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration Jefferson, AR, USA
| | - Jeffrey W Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration Jefferson, AR, USA
| | | |
Collapse
|
20
|
Hoermann R, Midgley JEM, Larisch R, Dietrich JW. Homeostatic Control of the Thyroid-Pituitary Axis: Perspectives for Diagnosis and Treatment. Front Endocrinol (Lausanne) 2015; 6:177. [PMID: 26635726 PMCID: PMC4653296 DOI: 10.3389/fendo.2015.00177] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022] Open
Abstract
The long-held concept of a proportional negative feedback control between the thyroid and pituitary glands requires reconsideration in the light of more recent studies. Homeostatic equilibria depend on dynamic inter-relationships between thyroid hormones and pituitary thyrotropin (TSH). They display a high degree of individuality, thyroid-state-related hierarchy, and adaptive conditionality. Molecular mechanisms involve multiple feedback loops on several levels of organization, different time scales, and varying conditions of their optimum operation, including a proposed feedforward motif. This supports the concept of a dampened response and multistep regulation, making the interactions between TSH, FT4, and FT3 situational and mathematically more complex. As a homeostatically integrated parameter, TSH becomes neither normatively fixed nor a precise marker of euthyroidism. This is exemplified by the therapeutic situation with l-thyroxine (l-T4) where TSH levels defined for optimum health may not apply equivalently during treatment. In particular, an FT3-FT4 dissociation, discernible FT3-TSH disjoint, and conversion inefficiency have been recognized in l-T4-treated athyreotic patients. In addition to regulating T4 production, TSH appears to play an essential role in maintaining T3 homeostasis by directly controlling deiodinase activity. While still allowing for tissue-specific variation, this questions the currently assumed independence of the local T3 supply. Rather it integrates peripheral and central elements into an overarching control system. On l-T4 treatment, altered equilibria have been shown to give rise to lower circulating FT3 concentrations in the presence of normal serum TSH. While data on T3 in tissues are largely lacking in humans, rodent models suggest that the disequilibria may reflect widespread T3 deficiencies at the tissue level in various organs. As a consequence, the use of TSH, valuable though it is in many situations, should be scaled back to a supporting role that is more representative of its conditional interplay with peripheral thyroid hormones. This reopens the debate on the measurement of free thyroid hormones and encourages the identification of suitable biomarkers. Homeostatic principles conjoin all thyroid parameters into an adaptive context, demanding a more flexible interpretation in the accurate diagnosis and treatment of thyroid dysfunction.
Collapse
Affiliation(s)
- Rudolf Hoermann
- Department of Nuclear Medicine, Klinikum Luedenscheid, Luedenscheid, Germany
| | | | - Rolf Larisch
- Department of Nuclear Medicine, Klinikum Luedenscheid, Luedenscheid, Germany
| | - Johannes W. Dietrich
- Medical Department I, Endocrinology and Diabetology, Bergmannsheil University Hospitals, Ruhr University of Bochum, Bochum, Germany
- Ruhr Center for Rare Diseases (CeSER), Ruhr University of Bochum and Witten/Herdecke University, Bochum, Germany
- *Correspondence: Johannes W. Dietrich,
| |
Collapse
|
21
|
Dietary high-fat lard intake induces thyroid dysfunction and abnormal morphology in rats. Acta Pharmacol Sin 2014; 35:1411-20. [PMID: 25263336 DOI: 10.1038/aps.2014.82] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 07/05/2014] [Indexed: 12/11/2022]
Abstract
AIM Excess dietary fat intake can induce lipotoxicity in non-adipose tissues. The aim of this study was to observe the effects of dietary high-fat lard intake on thyroid in rats. METHODS Male Sprague-Dawley rats were fed a high-fat lard diet for 24 weeks, and then the rats were fed a normal control diet (acute dietary modification) or the high-fat lard diet for another 6 weeks. The serum lipid profile, total thyroxine (TT4), free thyroxine (FT4) and thyrotropin (TSH) levels were determined at the 12, 18, 24 and 30 weeks. High-frequency ultrasound scanning of the thyroid glands was performed at the 24 or 30 weeks. After the rats were sacrificed, the thyroid glands were collected for histological and immunohistochemical analyses. RESULTS The high-fat lard diet significantly increased triglyceride levels in both the serum and thyroid, and decreased serum TT4 and FT4 levels in parallel with elevated serum TSH levels. Ultrasonic imaging revealed enlarged thyroid glands with lowered echotexture and relatively heterogeneous features in the high-fat lard fed rats. The thyroid glands from the high-fat lard fed rats exhibited enlarged follicle cavities and flattened follicular epithelial cells under light microscopy, and dilated endoplasmic reticulum cisternae, twisted nuclei, fewer microvilli and secretory vesicles under transmission electron microscopy. Furthermore, the thyroid glands from the high-fat lard fed rats showed markedly low levels of thyroid hormone synthesis-related proteins TTF-1 and NIS. Acute dietary modification by withdrawal of the high-fat lard diet for 6 weeks failed to ameliorate the high-fat lard diet-induced thyroid changes. CONCLUSION Dietary high-fat lard intake induces significant thyroid dysfunction and abnormal morphology in rats, which can not be corrected by short-term dietary modification.
Collapse
|
22
|
Ekerot P, Ferguson D, Glämsta EL, Nilsson LB, Andersson H, Rosqvist S, Visser SAG. Systems pharmacology modeling of drug-induced modulation of thyroid hormones in dogs and translation to human. Pharm Res 2013; 30:1513-24. [PMID: 23568527 DOI: 10.1007/s11095-013-0989-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 01/22/2013] [Indexed: 10/27/2022]
Abstract
PURPOSE To develop a systems pharmacology model based on hormone physiology and pharmacokinetic-pharmacodynamic concepts describing the impact of thyroperoxidase (TPO) inhibition on thyroid hormone homeostasis in the dog and to predict drug-induced changes in thyroid hormones in humans. METHODS A population model was developed based on a simultaneous analysis of concentration-time data of T₄, T₃ and TSH in dogs following once daily oral dosing for up to 6-months of a myeloperoxidase inhibitor (MPO-IN1) with TPO inhibiting properties. The model consisted of linked turnover compartments for T₄, T₃ and TSH including a negative feedback from T₄ on TSH concentrations. RESULTS The model could well describe the concentration-time profiles of thyroid hormones in dog. Successful model validation was performed by predicting the hormone concentrations during 1-month administration of MPO-IN2 based on its in vitro dog TPO inhibition potency. Using human thyroid hormone turnover rates and TPO inhibitory potency, the human T₄ and TSH concentrations upon MPO-IN1 treatment were predicted well. CONCLUSIONS The model provides a scientific framework for the prediction of drug induced effects on plasma thyroid hormones concentrations in humans via TPO inhibition based on results obtained in in vitro and animal studies.
Collapse
Affiliation(s)
- Petra Ekerot
- Modeling & Simulation, DMPK CNSP, AstraZeneca R&D, 15185 Södertälje, Sweden
| | | | | | | | | | | | | |
Collapse
|
23
|
Lumen A, Mattie DR, Fisher JW. Evaluation of Perturbations in Serum Thyroid Hormones During Human Pregnancy Due to Dietary Iodide and Perchlorate Exposure Using a Biologically Based Dose-Response Model. Toxicol Sci 2013; 133:320-41. [DOI: 10.1093/toxsci/kft078] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
24
|
Gilbert ME, Hedge JM, Valentín-Blasini L, Blount BC, Kannan K, Tietge J, Zoeller RT, Crofton KM, Jarrett JM, Fisher JW. An Animal Model of Marginal Iodine Deficiency During Development: The Thyroid Axis and Neurodevelopmental Outcome*. Toxicol Sci 2013; 132:177-95. [DOI: 10.1093/toxsci/kfs335] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
25
|
Fisher JW, Li S, Crofton K, Zoeller RT, McLanahan ED, Lumen A, Gilbert ME. Evaluation of Iodide Deficiency in the Lactating Rat and Pup Using a Biologically Based Dose-Response Model*. Toxicol Sci 2013; 132:75-86. [DOI: 10.1093/toxsci/kfs336] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
26
|
Zeise L, Bois FY, Chiu WA, Hattis D, Rusyn I, Guyton KZ. Addressing human variability in next-generation human health risk assessments of environmental chemicals. ENVIRONMENTAL HEALTH PERSPECTIVES 2013; 121:23-31. [PMID: 23086705 PMCID: PMC3553440 DOI: 10.1289/ehp.1205687] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 10/19/2012] [Indexed: 05/19/2023]
Abstract
BACKGROUND Characterizing variability in the extent and nature of responses to environmental exposures is a critical aspect of human health risk assessment. OBJECTIVE Our goal was to explore how next-generation human health risk assessments may better characterize variability in the context of the conceptual framework for the source-to-outcome continuum. METHODS This review was informed by a National Research Council workshop titled "Biological Factors that Underlie Individual Susceptibility to Environmental Stressors and Their Implications for Decision-Making." We considered current experimental and in silico approaches, and emerging data streams (such as genetically defined human cells lines, genetically diverse rodent models, human omic profiling, and genome-wide association studies) that are providing new types of information and models relevant for assessing interindividual variability for application to human health risk assessments of environmental chemicals. DISCUSSION One challenge for characterizing variability is the wide range of sources of inherent biological variability (e.g., genetic and epigenetic variants) among individuals. A second challenge is that each particular pair of health outcomes and chemical exposures involves combinations of these sources, which may be further compounded by extrinsic factors (e.g., diet, psychosocial stressors, other exogenous chemical exposures). A third challenge is that different decision contexts present distinct needs regarding the identification-and extent of characterization-of interindividual variability in the human population. CONCLUSIONS Despite these inherent challenges, opportunities exist to incorporate evidence from emerging data streams for addressing interindividual variability in a range of decision-making contexts.
Collapse
Affiliation(s)
- Lauren Zeise
- Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, Oakland, California 94612, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Dietrich JW, Landgrafe G, Fotiadou EH. TSH and Thyrotropic Agonists: Key Actors in Thyroid Homeostasis. J Thyroid Res 2012; 2012:351864. [PMID: 23365787 PMCID: PMC3544290 DOI: 10.1155/2012/351864] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 11/21/2012] [Indexed: 12/11/2022] Open
Abstract
This paper provides the reader with an overview of our current knowledge of hypothalamic-pituitary-thyroid feedback from a cybernetic standpoint. Over the past decades we have gained a plethora of information from biochemical, clinical, and epidemiological investigation, especially on the role of TSH and other thyrotropic agonists as critical components of this complex relationship. Integrating these data into a systems perspective delivers new insights into static and dynamic behaviour of thyroid homeostasis. Explicit usage of this information with mathematical methods promises to deliver a better understanding of thyrotropic feedback control and new options for personalised diagnosis of thyroid dysfunction and targeted therapy, also by permitting a new perspective on the conundrum of the TSH reference range.
Collapse
Affiliation(s)
- Johannes W. Dietrich
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| | - Gabi Landgrafe
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
- Klinik für Allgemein- und Visceralchirurgie, Agaplesion Bethesda Krankenhaus Wuppertal gGmbH, Hainstraße 35, 42109 Wuppertal, NRW, Germany
| | - Elisavet H. Fotiadou
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| |
Collapse
|
28
|
Mumtaz M, Fisher J, Blount B, Ruiz P. Application of physiologically based pharmacokinetic models in chemical risk assessment. J Toxicol 2012; 2012:904603. [PMID: 22523493 PMCID: PMC3317240 DOI: 10.1155/2012/904603] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 12/21/2011] [Indexed: 12/21/2022] Open
Abstract
Post-exposure risk assessment of chemical and environmental stressors is a public health challenge. Linking exposure to health outcomes is a 4-step process: exposure assessment, hazard identification, dose response assessment, and risk characterization. This process is increasingly adopting "in silico" tools such as physiologically based pharmacokinetic (PBPK) models to fine-tune exposure assessments and determine internal doses in target organs/tissues. Many excellent PBPK models have been developed. But most, because of their scientific sophistication, have found limited field application-health assessors rarely use them. Over the years, government agencies, stakeholders/partners, and the scientific community have attempted to use these models or their underlying principles in combination with other practical procedures. During the past two decades, through cooperative agreements and contracts at several research and higher education institutions, ATSDR funded translational research has encouraged the use of various types of models. Such collaborative efforts have led to the development and use of transparent and user-friendly models. The "human PBPK model toolkit" is one such project. While not necessarily state of the art, this toolkit is sufficiently accurate for screening purposes. Highlighted in this paper are some selected examples of environmental and occupational exposure assessments of chemicals and their mixtures.
Collapse
Affiliation(s)
- Moiz Mumtaz
- Computational Toxicology and Methods Development Laboratory, Division of Toxicology and Environmental Medicine (DTEM), Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA 30333, USA
| | - Jeffrey Fisher
- National Center for Toxicological Research, USFDA, Jefferson, AR 72079, USA
| | - Benjamin Blount
- Division of Laboratory Studies, National Center for Environmental Health, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30341, USA
| | - Patricia Ruiz
- Computational Toxicology and Methods Development Laboratory, Division of Toxicology and Environmental Medicine (DTEM), Agency for Toxic Substances and Disease Registry (ATSDR), Atlanta, GA 30333, USA
| |
Collapse
|
29
|
Fisher J, Lumen A, Latendresse J, Mattie D. Extrapolation of hypothalamic-pituitary-thyroid axis perturbations and associated toxicity in rodents to humans: case study with perchlorate. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2012; 30:81-105. [PMID: 22458857 DOI: 10.1080/10590501.2012.653889] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Functional aspects of the Hypothalamic-Pituitary-Thyroid (HPT) axis in rats and humans are compared, exposing why extrapolation of toxicant-induced perturbations in the rat HPT axis to the human HPT axis cannot be accomplished using default risk assessment methodology. Computational tools, such as biologically based dose response models for the HPT axis, are recommended to perform complex animal to human extrapolations involving the HPT axis. Experimental and computational evidence are presented that suggest perchlorate acts directly on the thyroid gland in rats. The apparent escape from perchlorate-induced inhibition of thyroidal uptake of radioactive iodide in humans is discussed along with "rebound" or increased thyroidal uptake of radioactive iodide observed after discontinued clinical treatment with perchlorate.
Collapse
Affiliation(s)
- Jeffrey Fisher
- Division of Biochemical Toxicology, National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR 72079, USA.
| | | | | | | |
Collapse
|
30
|
Marginal iodide deficiency and thyroid function: Dose–response analysis for quantitative pharmacokinetic modeling. Toxicology 2011; 283:41-8. [DOI: 10.1016/j.tox.2011.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 02/01/2011] [Accepted: 02/03/2011] [Indexed: 11/19/2022]
|
31
|
Nichols JW, Breen M, Denver RJ, Distefano JJ, Edwards JS, Hoke RA, Volz DC, Zhang X. Predicting chemical impacts on vertebrate endocrine systems. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2011; 30:39-51. [PMID: 20963851 DOI: 10.1002/etc.376] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Animals have evolved diverse protective mechanisms for responding to toxic chemicals of both natural and anthropogenic origin. From a governmental regulatory perspective, these protective responses complicate efforts to establish acceptable levels of chemical exposure. To explore this issue, we considered vertebrate endocrine systems as potential targets for environmental contaminants. Using the hypothalamic-pituitary-thyroid (HPT), hypothalamic-pituitary-gonad (HPG), and hypothalamic-pituitary-adrenal (HPA) axes as case examples, we identified features of these systems that allow them to accommodate and recover from chemical insults. In doing so, a distinction was made between effects on adults and those on developing organisms. This distinction was required because endocrine system disruption in early life stages may alter development of organs and organ systems, resulting in permanent changes in phenotypic expression later in life. Risk assessments of chemicals that impact highly regulated systems must consider the dynamics of these systems in relation to complex environmental exposures. A largely unanswered question is whether successful accommodation to a toxic insult exerts a fitness cost on individual animals, resulting in adverse consequences for populations. Mechanistically based mathematical models of endocrine systems provide a means for better understanding accommodation and recovery. In the short term, these models can be used to design experiments and interpret study findings. Over the long term, a set of validated models could be used to extrapolate limited in vitro and in vivo testing data to a broader range of untested chemicals, species, and exposure scenarios. With appropriate modification, Tier 2 assays developed in support of the U.S. Environmental Protection Agency's Endocrine Disruptor Screening Program could be used to assess the potential for accommodation and recovery and inform the development of mechanistically based models.
Collapse
Affiliation(s)
- John W Nichols
- U.S. Environmental Protection Agency, Duluth, Minnesota, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Kitchin KT, Conolly R. Arsenic-induced carcinogenesis--oxidative stress as a possible mode of action and future research needs for more biologically based risk assessment. Chem Res Toxicol 2010; 23:327-35. [PMID: 20035570 DOI: 10.1021/tx900343d] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Exposure to inorganic arsenic (iAs) induces cancer in human lungs, urinary bladder, skin, kidney, and liver, with the majority of deaths from lung and bladder cancer. To date, cancer risk assessments for iAs have not relied on mechanistic data, as we have lacked sufficient understanding of arsenic's pharmacokinetics and mode(s) of carcinogenic action (MOA). Furthermore, while there are vast amounts of toxicological data on iAs, relatively little of it has been collected using experimental designs that efficiently support development of biologically based dose-response (BBDR) models and subsequently risk assessment. This review outlines an efficient approach to the development of a BBDR model for iAs that would reduce uncertainties in its cancer risk assessment. This BBDR-based approach is illustrated by using oxidative stress as the carcinogenic MOA for iAs but would be generically applicable to other MOAs. Six major research needs that will facilitate BBDR model development for arsenic-induced cancer are (1) MOA research, which is needed to reduce the uncertainty in risk assessment; (2) development and integration of the pharmacodynamic component (MOA) of the BBDR model; (3) dose-response and extrapolation model selection; (4) the determination of internal human speciated arsenical concentrations to improve physiologically based pharmacokinetic (PBPK) models; (5) animal models of arsenic carcinogenesis; and (6) the determination of the low dose human relationship for death from cancer, particularly in lungs and urinary bladder. The major parts of the BBDR model are arsenic exposure, a physiologically based pharmacokinetic model, reactive species, antioxidant defenses, oxidative stress, cytotoxicity, growth factors, transcription factors, DNA damage, chromosome damage, cell proliferation, mutation accumulation, and cancer. The BBDR model will need to be developed concurrently with data collection so that model uncertainties can be identified and addressed through an iterative process of targeted additional research.
Collapse
Affiliation(s)
- Kirk T Kitchin
- Mail Drop B143-06, Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA.
| | | |
Collapse
|
33
|
McLanahan ED, Andersen ME, Campbell JL, Fisher JW. Competitive inhibition of thyroidal uptake of dietary iodide by perchlorate does not describe perturbations in rat serum total T4 and TSH. ENVIRONMENTAL HEALTH PERSPECTIVES 2009; 117:731-8. [PMID: 19479014 PMCID: PMC2685834 DOI: 10.1289/ehp.0800111] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Accepted: 01/05/2009] [Indexed: 05/03/2023]
Abstract
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.
Collapse
Affiliation(s)
- Eva D. McLanahan
- University of Georgia, Department of Environmental Health Sciences, Athens, Georgia, USA
| | - Melvin E. Andersen
- Hamner Institutes for Health Sciences, Division of Computational Biology, Research Triangle Park, North Carolina, USA
| | - Jerry L. Campbell
- University of Georgia, Department of Environmental Health Sciences, Athens, Georgia, USA
| | - Jeffrey W. Fisher
- University of Georgia, Department of Environmental Health Sciences, Athens, Georgia, USA
- Address correspondence to J.W. Fisher, 206 Environmental Health Sciences Department, University of Georgia, Athens, GA 30602-2102 USA. Telephone: (706) 542-1001. Fax: (706) 542-7472. E-mail:
| |
Collapse
|