Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 202
1.
Physiol Rep ; 12(8): e16007, 2024 Apr.
Article En | MEDLINE | ID: mdl-38658325

Thyroid hormones regulate metabolic rate, nutrient utilization, growth, and development. Swine are susceptible to thyroid suppression in response to disease or environmental conditions, but the physiological impact of such disruption has not been established. The objective of this study was to evaluate the impact of hypothyroidism induced with the antithyroid medication methimazole (MMI). 10 mg/kg MMI significantly decreased circulating triiodothyronine (T3) for the duration of treatment but had only a transient effect on circulating thyroxine (T4). Thyroid tissue weight was significantly increased by more than 3.5-fold in response to MMI treatment. Histologically, the eosinophilic colloid was largely absent from the thyroid follicle which displayed a disorganized columnar epithelium consistent with goiter. MMI induced hypothyroidism has no effect on growth rate over 28 days. Hepatic expression of genes associated with thyroid metabolism (DIO1, DIO2, and DIO3), lipid utilization (CD36, FASN, and ACACA), apoptosis (TP53, PERP, SIVA1, and SFN) and proliferation (CDK1, CDK2, CDK4, and CDKN1A) were unaffected by treatment. Collectively these results demonstrate that MMI induces mild systemic hypothyroidism and pronounced goiter, indicating a strong homeostatic central regulation within the hypothalamic pituitary thyroid axis. This combined with limited peripheral effects, indicates resilience to hypothyroidism in modern swine.


Antithyroid Agents , Hypothyroidism , Methimazole , Thyroid Gland , Animals , Methimazole/toxicity , Methimazole/adverse effects , Hypothyroidism/chemically induced , Hypothyroidism/metabolism , Swine , Antithyroid Agents/toxicity , Antithyroid Agents/adverse effects , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Gland/pathology , Female , Triiodothyronine/blood , Liver/metabolism , Liver/drug effects , Liver/pathology , Thyroxine/blood , Male
2.
J Proteomics ; 274: 104811, 2023 03 15.
Article En | MEDLINE | ID: mdl-36592655

The central goal of this study was to investigate the alterations in transcriptome of testis in F1 generation adult rats exposed to carbimazole prenatally. At post-natal day 100, the testis of rats delivered to carbimazole exposed (time-mated pregnant rats orally administered with carbimazole from gestation day 9 to 21) and control (untreated pregnant rats) groups were subjected to transcriptomic analysis using NGS platform. A total of 187 differentially expressed (up regulated: 49 genes; down regulated: 138) genes were identified in carbimazole exposed rats over controls and the major processes associated with these altered testicular transcripts were examined. Functional clustering analysis suggest that the involvement of identified DEGs were linked to intrinsic and extrinsic apoptotic pathways, mitochondrial solute carriers slc25a members, nuclear receptors/zinc family members, steroidogenic pathway and cholesterol synthesis, and growth factors and protein kinases and thus represent potential mediators of the developmental toxic effects of carbimazole in F1 generation rats. Based on the findings, it can be concluded that prenatal exposure to carbimazole prominently affects expression of multiple transcripts implicating key regulatory events associated with testicular functions, spermatogenesis and steroidogenesis in rats at their adulthood. These results support our earlier findings and hypothesis. This background information obtained at the testicular transcriptome during gestational hypothyroidism might be helpful for future studies and experiments to gain additional in-depth analysis and to develop strategies to protect F1 generation male reproductive health. SIGNIFICANCE: The rationale for the paper described thyroid gland changes in the off springs. Antithyroid drugs are widely used to control thyroid disorders and used to control thyroid hormone levels during surgeries. Carbimazole is one of the antithyroid drugs and is a parent molecule of methimazole. Both the drugs can able to cross placenta. During fetal period, the development of thyroid gland is not completely formed and hence, the fetus entirely depends on the maternal thyroid hormones. Therefore, it is conceivable that the disturbances at the level of maternal thyroid hormones could interfere with the development of vital organs such as testis and glands including thyroid gland (Kala et al., 2012). To address this notion, the present study was designed and executed.


Antithyroid Agents , Carbimazole , Pregnancy , Female , Rats , Male , Animals , Carbimazole/metabolism , Carbimazole/pharmacology , Antithyroid Agents/toxicity , Transcriptome , Testis/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology
3.
Aquat Toxicol ; 244: 106094, 2022 Mar.
Article En | MEDLINE | ID: mdl-35134604

Several chemicals have been identified as thyroid hormone axis disrupting chemicals (THADCs) able to interfere with the thyroid hormone system during fetal life and early life stages, thereby impairing neurodevelopment in mammals and inducing development and growth disorders in fish and amphibians. However, identification of THADCs is particularly challenging, and thyroid modalities are currently only assessed in vivo by mammalian and amphibian tests. The aquatic African clawed frog (Xenopus laevis/tropicalis) is the model species of the amphibian test guidelines developed by the OECD and the United States Environmental Protection Agency, but as most European amphibians are semi-aquatic, concern has been raised whether the sensitivity of native European species is comparable to Xenopus. A shortened version of the OEDC test guideline 241 (Larval Amphibian Growth and Development Assay, LAGDA) was used to investigate the effects of two model THADCs on the metamorphosis and thyroid histopathology in the European common frog (Rana temporaria). R. temporaria eggs were collected on the field and exposed till metamorphic climax to sodium perchlorate (11.9-426.5 µg/L perchlorate concentrations) and 6-propylthiouracil (PTU: 1.23-47.7 mg/L). PTU severely delayed metamorphosis and affected several thyroid gland histopathological endpoints at slightly lower concentrations compared to Xenopus. As opposed to what was described in similar Xenopus studies, we observed no effect of perchlorate on the investigated endpoints. Interspecies differences may be linked to mechanisms of action.


Propylthiouracil , Water Pollutants, Chemical , Animals , Antithyroid Agents/toxicity , Larva , Metamorphosis, Biological , Perchlorates/toxicity , Propylthiouracil/toxicity , Rana temporaria , Sodium Compounds , Thyroid Gland , Water Pollutants, Chemical/toxicity , Xenopus laevis
4.
Toxicol Lett ; 354: 44-55, 2022 Jan 01.
Article En | MEDLINE | ID: mdl-34757178

Disruption of the thyroid hormone system during development can impair brain development and cause irreversible damage. Some thyroid hormone system disruptors act by inhibiting the thyroperoxidase (TPO) enzyme, which is key to thyroid hormone synthesis. For the potent TPO-inhibiting drug propylthiouracil (PTU) this has been shown to result in thyroid hormone system disruption and altered brain development in animal studies. However, an outstanding question is which chemicals beside PTU can cause similar effects on brain development and to what degree thyroid hormone insufficiency must be induced to be able to measure adverse effects in rats and their offspring. To start answering these questions, we performed a perinatal exposure study in pregnant rats with two TPO-inhibitors: the drug methimazole (MMI) and the triazole herbicide amitrole. The study involved maternal exposure from gestational day 7 through to postnatal day 22, to MMI (8 and 16 mg/kg body weight/day) or amitrole (25 and 50 mg/kg body weight/day). Both MMI and amitrole reduced serum T4 concentrations in a dose-dependent manner in dams and offspring, with a strong activation of the hypothalamic-pituitary-thyroid axis. This reduction in serum T4 led to decreased thyroid hormone-mediated gene expression in the offspring's brains and caused adverse effects on brain function, seen as hyperactivity and decreased habituation in preweaning pups. These dose-dependent effects induced by MMI and amitrole are largely the same as those observed with PTU. This demonstrates that potent TPO-inhibitors can induce effects on brain development in rats and that these effects are driven by T4 deficiency. This knowledge will aid the identification of TPO-inhibiting thyroid hormone system disruptors in a regulatory context and can serve as a starting point in search of more sensitive markers of developmental thyroid hormone system disruption.


Amitrole/toxicity , Antithyroid Agents/toxicity , Enzyme Inhibitors/toxicity , Methimazole/toxicity , Motor Activity/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Thyroid Gland/drug effects , Animals , Animals, Newborn , Disease Models, Animal , Female , Maternal Exposure/adverse effects , Neurotoxicity Syndromes/physiopathology , Pregnancy , Rats , Signal Transduction/drug effects , Thyroid Function Tests
5.
Environ Toxicol Pharmacol ; 87: 103738, 2021 Oct.
Article En | MEDLINE | ID: mdl-34492396

The effects of two drugs containing the synthetic thyroid hormone levothyroxine (LEV) and an anti-thyroid drug containing propylthiouracil (PTU) on the three early life stages of Xenopus laevis were evaluated with the Frog Embryo Teratogenesis Assay-Xenopus, Tadpole Toxicity Test, and Amphibian Metamorphosis Assay using biochemical and morphological markers. Tested drugs caused more effective growth retardation in stage 8 embryos than stage 46 tadpoles. Significant inhibition of biomarker enzymes has been identified in stage 46 tadpoles for both drugs. AMA test results showed that LEV-I caused progression in the developmental stage and an increase in thyroxine level in 7 days exposure and growth retardation in 21 days exposure in stage 51 tadpoles. On the other hand, increases in lactate dehydrogenase activity for both drugs in the AMA test may be due to impacted energy metabolism during sub-chronic exposure. These results also show that the sensitivity and responses of Xenopus laevis at different early developmental stages may be different when exposed to drugs.


Antithyroid Agents/toxicity , Embryo, Nonmammalian/drug effects , Larva/drug effects , Propylthiouracil/toxicity , Teratogens/toxicity , Thyroxine/toxicity , Xenopus laevis , Acetylcholinesterase/metabolism , Animals , Carboxylesterase/metabolism , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/enzymology , Embryonic Development/drug effects , Female , Glutathione Reductase/metabolism , Glutathione Transferase/metabolism , Larva/enzymology , Larva/growth & development , Male , Metamorphosis, Biological/drug effects , Xenopus laevis/abnormalities , Xenopus laevis/growth & development , Xenopus laevis/metabolism
6.
Diabetologia ; 63(9): 1822-1835, 2020 09.
Article En | MEDLINE | ID: mdl-32472193

AIMS/HYPOTHESIS: During pregnancy, maternal metabolic disease and hormonal imbalance may alter fetal beta cell development and/or proliferation, thus leading to an increased risk for developing type 2 diabetes in adulthood. Although thyroid hormones play an important role in fetal endocrine pancreas development, the impact of maternal hypothyroidism on glucose homeostasis in adult offspring remains poorly understood. METHODS: We investigated this using a mouse model of hypothyroidism, induced by administration of an iodine-deficient diet supplemented with propylthiouracil during gestation. RESULTS: Here, we show that, when fed normal chow, adult mice born to hypothyroid mothers were more glucose-tolerant due to beta cell hyperproliferation (two- to threefold increase in Ki67-positive beta cells) and increased insulin sensitivity. However, following 8 weeks of high-fat feeding, these offspring gained 20% more body weight, became profoundly hyperinsulinaemic (with a 50% increase in fasting insulin concentration), insulin-resistant and glucose-intolerant compared with controls from euthyroid mothers. Furthermore, altered glucose metabolism was maintained in a second generation of animals. CONCLUSIONS/INTERPRETATION: Therefore, gestational hypothyroidism induces long-term alterations in endocrine pancreas function, which may have implications for type 2 diabetes prevention in affected individuals.


Blood Glucose/metabolism , Glucose Intolerance/metabolism , Hypothyroidism/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/embryology , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Antithyroid Agents/toxicity , Cell Proliferation , Diet, High-Fat , Disease Models, Animal , Female , Hyperinsulinism/metabolism , Insulin Resistance , Iodine/deficiency , Islets of Langerhans/metabolism , Mice , Pregnancy , Propylthiouracil/toxicity , Stress, Physiological
7.
Biotech Histochem ; 95(7): 522-531, 2020 Oct.
Article En | MEDLINE | ID: mdl-32124645

We investigated the effects of melatonin on rats with induced hypothyroidism during gestation as well as its effect on the development of the gonads of their offspring. Fifteen pregnant rats were divided into three groups: GC, rats without induced hypothyroidism; GH, rats with induced hypothyroidism; GHM, rats with induced hypothyroidism plus melatonin. Hypothyroidism was induced by oral administration of 6-propyl-2-thiouracil and melatonin was applied subcutaneously. Treatments were performed during gestation and lactation. For the matrices, we evaluated the number of pups, body weight gain, ovarian weight, thyroid weight, organosomatic index, thyroid stimulating hormone (TSH) dose and thyroid morphometry. For the pups, weight gain, TSH, weight, morphometry of the gonads and organosomatic index were analyzed, as well as the cell proliferation index. TSH was elevated only in the matrices of GH animals. Melatonin prevented reduction of ovarian and thyroid weight, number of pups, follicular diameter and thyroid epithelial proportion of the matrices with hypothyroidism. The offspring of rats of the GH group exhibited less body weight gain, gonad and thyroid weight, and gonad cell proliferation index compared to the offspring born of rats of the GC and GHM groups. Melatonin prevented the effects of maternal hypothyroidism on the offspring of rats.


Gonads/drug effects , Hypothyroidism/chemically induced , Melatonin/pharmacology , Pregnancy Complications/chemically induced , Thyroid Gland/drug effects , Animals , Antioxidants/pharmacology , Antithyroid Agents/toxicity , Female , Gonads/growth & development , Pregnancy , Prenatal Exposure Delayed Effects , Propylthiouracil/toxicity , Rats , Thyroid Gland/growth & development
8.
Reprod Biol ; 20(2): 247-253, 2020 Jun.
Article En | MEDLINE | ID: mdl-32089504

Hypothyroidism affects the content of triacylglycerol (TAG), total cholesterol (TC), oxidized lipids, glycogen, and infiltration of immune cells into the ovary and uterus. This study aimed to analyze the impact of hypothyroidism on the lipid content of different regions of the oviduct. Control (n = 6) and hypothyroid (n = 6; 10 mg/kg/day of methimazole in the drinking water for 30 days) adult rabbits were used. In the fimbriae/infundibulum (FIM/INF), ampulla, (AMP), isthmus (IST), and utero-tubal junction (UTJ), the TAG and TC concentrations, presence of oxidized lipid, relative expressions of perilipin A (PLIN A), peroxisome proliferator-activated receptor γ (PPARγ), CAAT/enhancer-binding protein α (C/EBPα), and farnesoid X receptor (FXRα) were analyzed. The content of glycogen and glycans, as well as the infiltration of lymphocytes, were also quantified. In the FIM/INF, hypothyroidism reduced the content of TC, expression of C/EBPα, and presence of glycans while increased the number of intraepithelial lymphocytes. In the AMP and IST-UTJ regions, hypothyroidism increased the content of TAG, oxidized lipids, expression of PPARγ, and glycogen content but decreased the expression of PLIN-A. The FXRα expression in secretory cells of IST-UTJ was higher in the hypothyroid rabbits compared to controls. Additionally, hypothyroidism reduced the C/EBPα expression and the number of intraepithelial lymphocytes in the AMP and IST-UTJ regions, respectively. We demonstrated that the effect of hypothyroidism depends on the oviductal region, possibly associated with different physiological functions specific to each region. These alterations may be related to infertility, tubal disturbances, and ectopic pregnancy observed in hypothyroid women.


Fallopian Tubes/cytology , Glycogen/chemistry , Hypothyroidism/veterinary , Lipids/chemistry , Lymphocytes/physiology , Rabbits , Animals , Antithyroid Agents/toxicity , Female , Glycogen/metabolism , Hypothyroidism/chemically induced , Lipid Metabolism , Methimazole/toxicity
9.
Ecotoxicol Environ Saf ; 188: 109875, 2020 Jan 30.
Article En | MEDLINE | ID: mdl-31706244

Previous works showed that chronic exposure to Aroclor 1254 disrupted glucose homeostasis and induced insulin resistance in male mice. To further observe the different effects of Aroclor 1254 exposure on the pancreatic α-cells and ß-cells, male mice were exposed to Aroclor 1254 (0, 0.5, 5, 50, 500 µg/kg) for 60 days, the pancreas was performed a histological examination. The results showed that the percentage of apoptosis cell (indicated by TUNEL assay) was increased in both α-cells and ß-cells, as the Aroclor 1254 dose was increased; the proliferation (indicated by PCNA expression) rate of ß-cells was elevated while that of α-cells was not affected, resulting in an increased ß-cell mass and a decreased α-cell mass in a dose-depend manner. The number of Pdx-1 positive ß-cells was significantly increased whereas that of Arx positive α-cells was markedly decreased, indicating an enhanced ß-cell neogenesis and a weakened α-cell neogenesis. The drastically reduction of serum testosterone levels in all the treatments suggested an anti-androgenic potency of Aroclor 1254. The up-regulation of estrogen receptors (ERα and ERß) and androgen receptor in ß-cells might be responsible for the increased ß-cell mass and neogenesis.


Antithyroid Agents/toxicity , Glucagon-Secreting Cells/drug effects , Insulin-Secreting Cells/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Testosterone/blood , Trans-Activators/metabolism , Transcription Factors/metabolism
10.
Thyroid ; 29(10): 1425-1437, 2019 10.
Article En | MEDLINE | ID: mdl-31298630

Background: Thyroid tumor progression from well-differentiated cancer to poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) involves step-wise dedifferentiation associated with loss of iodine avidity and poor outcomes. ALK fusions, typically STRN-ALK, are found with higher incidence in human PDTC compared with well-differentiated cancer and, as previously shown, can drive the development of murine PDTC. The aim of this study was to evaluate thyroid cancer initiation and progression in mice with concomitant expression of STRN-ALK and inactivation of the tumor suppressor p53 (Trp53) in thyroid follicular cells. Methods: Transgenic mice with thyroid-specific expression of STRN-ALK and biallelic p53 loss were generated and aged on a regular diet or with methimazole and sodium perchlorate goitrogen treatment. Development and progression of thyroid tumors were monitored by using ultrasound imaging, followed by detailed histological and immunohistochemical evaluation. Gene expression analysis was performed on selected tumor samples by using RNA-Seq and quantitative RT-PCR. Results: In mice treated with goitrogen, the first thyroid cancers appeared at 6 months of age, reaching 86% penetrance by the age of 12 months, while a similar rate (71%) of tumor occurrence in mice on regular diet was observed by 18 months of age. Histological examination revealed well-differentiated papillary thyroid carcinomas (PTC) (n = 26), PDTC (n = 21), and ATC (n = 8) that frequently coexisted in the same thyroid gland. The tumors were frequently lethal and associated with the development of lung metastasis in 24% of cases. Histological and immunohistochemical characteristics of these cancers recapitulated tumors seen in humans. Detailed analysis of PDTC revealed two tumor types with distinct cell morphology and immunohistochemical characteristics, designated as PDTC type 1 (PDTC1) and type 2 (PDTC2). Gene expression analysis showed that PDTC1 tumors retained higher expression of thyroid differentiation genes including Tg and Slc5a5 (Nis) as compared with PDTC2 tumors. Conclusions: In this study, we generated a new mouse model of multistep thyroid cancer dedifferentiation with evidence of progression from PTC to PDTC and ATC. Further, PDTC in these mice showed two distinct histologic appearances correlated with levels of expression of thyroid differentiation and iodine metabolism genes, suggesting a possibility of existence of two PDTC types with different functional characteristics and potential implication for therapeutic approaches to these tumors.


Anaplastic Lymphoma Kinase/genetics , Calmodulin-Binding Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Oncogene Proteins, Fusion/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Animals , Antithyroid Agents/toxicity , Cell Dedifferentiation/genetics , Cell Differentiation/genetics , Disease Models, Animal , Disease Progression , Methimazole/toxicity , Mice , Mice, Knockout , Mice, Transgenic , Perchlorates/toxicity , RNA-Seq , Sodium Compounds/toxicity , Symporters/genetics , Thyroglobulin/genetics , Thyroid Cancer, Papillary/chemically induced , Thyroid Cancer, Papillary/genetics , Thyroid Carcinoma, Anaplastic/chemically induced , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Neoplasms/chemically induced , Thyroid Neoplasms/genetics , Transcriptome
11.
Environ Monit Assess ; 191(7): 454, 2019 Jun 21.
Article En | MEDLINE | ID: mdl-31222463

It is well-known that the metamorphosis process in amphibians is dependent on thyroid hormones. Laboratory studies have shown that several environmental contaminants can affect the function of thyroid hormones leading to alterations in the amphibian metamorphosis. The basic idea of the present study was to elucidate if the amphibian metamorphosis might be a useful tool as biomarker for effect-based environmental monitoring, examining wild tadpoles for potential thyroid hormone disruption. A laboratory test was performed to identify the responses from exposure to 6-propylthiouracil (PTU), which has a well-known mechanism on the thyroid system, on Swedish tadpoles from the Rana genus. This was followed by an environmental monitoring study where tadpoles of Rana arvalis, R. temporaria, and Bufo bufo were sampled from various sites in Sweden. Morphological data such as body weight, histopathological measurements of the thyroid glands, and environmental parameters were recorded. The results revealed that Rana tadpoles respond similar as other amphibians to PTU exposure, with interrupted development and increased size relative to the developmental stage. Data on some wild tadpoles showed similar features as the PTU exposed, such as high body weight, thus suggesting potential thyroid disrupting effects. However, histological evaluation of thyroid glands and pesticide analyses of the water revealed no clear evidence of chemical interactions. To a minor degree, the changes in body weight may be explained by natural circumstances such as pH, forest cover, and temperature. The present study cannot fully explain whether the high body weights recorded in some tadpoles have natural or chemical explanations. However, the study reveals that it is clearly achievable to catch tadpoles in suitable stages for the use in this type of biomonitoring and that the use of these biomarkers for assessment of thyroid disruption seems to be highly relevant.


Antithyroid Agents/toxicity , Bufonidae/embryology , Environmental Monitoring/methods , Larva/growth & development , Pesticides/toxicity , Propylthiouracil/toxicity , Ranidae/embryology , Thyroid Gland/pathology , Animals , Metamorphosis, Biological , Sweden , Thyroid Gland/drug effects , Thyroid Hormones/metabolism
12.
Thyroid ; 29(7): 934-945, 2019 07.
Article En | MEDLINE | ID: mdl-31084419

Background: Hypothyroidism, the most common endocrine disease, induces cardiac electrical remodeling that creates a substrate for ventricular arrhythmias. Recent studies report that high thyrotropin (TSH) levels are related to cardiac electrical abnormalities and increased mortality rates. The aim of the present work was to investigate the direct effects of TSH on the heart and its possible causative role in the increased incidence of arrhythmia in hypothyroidism. Methods: A new rat model of central hypothyroidism (low TSH levels) was created and characterized together with the classical propylthiouracil-induced primary hypothyroidism model (high TSH levels). Electrocardiograms were recorded in vivo, and ionic currents were recorded from isolated ventricular myocytes in vitro by the patch-clamp technique. Protein and mRNA were measured by Western blot and quantitative reverse transcription polymerase chain reaction in rat and human cardiac myocytes. Adult human action potentials were simulated in silico to incorporate the experimentally observed changes. Results: Both primary and central hypothyroidism models increased the L-type Ca2+ current (ICa-L) and decreased the ultra-rapid delayed rectifier K+ current (IKur) densities. However, only primary but not central hypothyroidism showed electrocardiographic repolarization abnormalities and increased ventricular arrhythmia incidence during caffeine/dobutamine challenge. These changes were paralleled by a decrease in the density of the transient outward K+ current (Ito) in cardiomyocytes from animals with primary but not central hypothyroidism. In vitro treatment with TSH for 24 hours enhanced isoproterenol-induced spontaneous activity in control ventricular cells and diminished Ito density in cardiomyocytes from control and central but not primary hypothyroidism animals. In human myocytes, TSH decreased the expression of KCND3 and KCNQ1, Ito, and the delayed rectifier K+ current (IKs) encoding proteins in a protein kinase A-dependent way. Transposing the changes produced by hypothyroidism and TSH to a computer model of human ventricular action potential resulted in enhanced occurrence of early afterdepolarizations and arrhythmia mostly in primary hypothyroidism, especially under ß-adrenergic stimulation. Conclusions: The results suggest that suppression of repolarizing K+ currents by TSH underlies most of the electrical remodeling observed in hypothyroidism. This work demonstrates that the activation of the TSH-receptor/protein kinase A pathway in the heart is responsible for the cardiac electrical remodeling and arrhythmia generation seen in hypothyroidism.


Arrhythmias, Cardiac/metabolism , Atrial Remodeling/physiology , Hypothyroidism/metabolism , Myocytes, Cardiac/metabolism , Thyrotropin/metabolism , Action Potentials , Animals , Antithyroid Agents/toxicity , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Bexarotene/toxicity , Calcium/metabolism , Computer Simulation , Disease Models, Animal , Disease Susceptibility , Electrocardiography , Humans , Hypothyroidism/complications , Hypothyroidism/physiopathology , Isoproterenol/pharmacology , KCNQ1 Potassium Channel/drug effects , KCNQ1 Potassium Channel/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Propylthiouracil/toxicity , RNA, Messenger/metabolism , Rats , Shal Potassium Channels/drug effects , Shal Potassium Channels/genetics , Thyrotropin/pharmacology
13.
Neurotoxicology ; 74: 47-57, 2019 09.
Article En | MEDLINE | ID: mdl-31121238

High throughput in vitro, in silico, and computational approaches have identified numerous environmental chemicals that interfere with thyroid hormone (TH) activity, and it is posited that human exposures to such chemicals are a contributing factor to neurodevelopmental disorders. However, whether hits in screens of TH activity are predictive of developmental neurotoxicity (DNT) has yet to be systematically addressed. The zebrafish has been proposed as a second tier model for assessing the in vivo DNT potential of TH active chemicals. As an initial evaluation of the feasibility of this proposal, we determined whether an endpoint often used to assess DNT in larval zebrafish, specifically photomotor behavior, is altered by experimentally induced hyper- and hypothyroidism. Developmental hyperthyroidism was simulated by static waterborne exposure of zebrafish to varying concentrations (3-300 nM) of thyroxine (T4) or triiodothyronine (T3) beginning at 6 h post-fertilization (hpf) and continuing through 5 days post-fertilization (dpf). Teratogenic effects and lethality were observed at 4 and 5 dpf in fish exposed to T4 or T3 at concentrations >30 nM. However, as early as 3 dpf, T4 (> 3 nM) and T3 (> 10 nM) significantly increased swimming activity triggered by sudden changes from light to dark, particularly during the second dark period (Dark 2). Conversely, developmental hypothyroidism, which was induced by treatment with 6-propyl-2-thiouracil (PTU), morpholino knockdown of the TH transporter mct8, or ablation of thyroid follicles in adult females prior to spawning, generally decreased swimming activity during dark periods, although effects did vary across test days. All effects of developmental hypothyroidism on photomotor behavior occurred independent of teratogenic effects and were most robust during Dark 2. Treatment with the T4 analog, Tetrac, restored photomotor response in mct8 morphants to control levels. Collectively, these findings suggest that while the sensitivity of photomotor behavior in larval zebrafish to detect TH disruption is influenced by test parameters, this test can distinguish between TH promoting and TH blocking activity and may be useful for assessing the DNT potential of TH-active chemicals.


Motor Activity/drug effects , Thyroid Hormones/toxicity , Animals , Antithyroid Agents/toxicity , Embryo, Nonmammalian , Female , Hyperthyroidism/chemically induced , Hyperthyroidism/psychology , Hypothyroidism/chemically induced , Hypothyroidism/psychology , Larva , Male , Monocarboxylic Acid Transporters/biosynthesis , Monocarboxylic Acid Transporters/genetics , Neurotoxicity Syndromes/psychology , Photic Stimulation , Swimming , Teratogens/toxicity , Thyroxine/blood , Thyroxine/toxicity , Triiodothyronine/blood , Triiodothyronine/toxicity , Zebrafish
14.
J Appl Toxicol ; 39(9): 1248-1256, 2019 09.
Article En | MEDLINE | ID: mdl-31066086

Indoor dust contains a multitude of industrial chemicals, and ingestion of dust is considered an important exposure route to organic contaminants. Some of these contaminants have been shown to interfere with the thyroid system, which may result in significant consequences on public health. The amphibian metamorphosis is a thyroid hormone-dependent process, which can be used as an in vivo model for studies on thyroid hormone-disrupting potency. Three contaminants of indoor dust were tested on metamorphosing Silurana (Xenopus) tropicalis tadpoles. The tested chemicals were Tris (1,3-dichloroisopropyl) phosphate (TDCiPP), tetrabromobisphenol-A (TBBPA) and propylparaben (PrP). Measurements reflecting general growth, development progress and thyroid epithelial cell height were performed on the exposed tadpoles as well as chemical analyses of the exposure water. It was shown that TDCiPP acts as a thyroid hormone-disrupting chemical in metamorphosing tadpoles by causing increased epithelial cell height in thyroid glands after exposure to a nominal concentration of 0.010 mg/L and in higher concentrations. TBBPA caused reductions in general growth of tadpoles at the nominal concentration 0.125 mg/L, and PrP caused acute toxicity at the nominal concentration 12.5 mg/L. However, no evident indications of specific thyroid-disrupting effects caused by TBBPA or PrP were observed.


Air Pollutants/chemistry , Air Pollutants/toxicity , Antithyroid Agents/toxicity , Dust/analysis , Larva/drug effects , Metamorphosis, Biological/drug effects , Thyroid Gland/drug effects , Xenopus/growth & development , Animals , Environmental Monitoring/methods , Models, Animal
15.
Drug Chem Toxicol ; 42(6): 615-623, 2019 Nov.
Article En | MEDLINE | ID: mdl-29722569

Drug-induced liver injury is a major concern in clinical studies as well as in post-marketing surveillance. Previous evidence suggested that drug exposure during periods of inflammation could increase an individual's susceptibility to drug hepatoxicity. The antithyroid drugs, methimazole (MMI) and propylthiouracil (PTU) can cause adverse reactions in patients, with liver as a usual target. We tested the hypothesis that MMI and PTU could be rendered hepatotoxic in animals undergoing a modest inflammation. Mice were treated with a nonhepatotoxic dose of LPS (100 µg/kg, i.p) or its vehicle. Nonhepatotoxic doses of MMI (10, 25 and 50 mg/kg, oral) and PTU (10, 25 and 50 mg/kg, oral) were administered two hours after LPS treatment. It was found that liver injury was evident only in animals received both drug and LPS, as estimated by increases in serum alanine aminotransferase (ALT), lactate dehydrogenase (LDH), aspartate aminotransferase (AST), and TNF-α. An increase in liver myeloperoxidase (MPO) enzyme activity and tissue lipid peroxidation (LPO) in addition of liver glutathione (GSH) depletion were also detected in LPS and antithyroid drugs cotreated animals. Furthermore, histopathological changes including, endotheliitis, fatty changes, severe inflammatory cells infiltration (hepatitis) and sinusoidal congestion were detected in liver tissue. Methyl palmitate (2 g/kg, i.v, 44 hours before LPS), as a macrophage suppressor, significantly alleviated antithyroids hepatotoxicity in LPS-treated animals. The results indicate a synergistic liver injury from antithyroid drugs and bacterial lipopolysaccharide coexposure.


Antithyroid Agents/toxicity , Chemical and Drug Induced Liver Injury/etiology , Inflammation/complications , Methimazole/toxicity , Propylthiouracil/toxicity , Animals , Antithyroid Agents/administration & dosage , Chemical and Drug Induced Liver Injury/pathology , Dose-Response Relationship, Drug , Lipopolysaccharides/toxicity , Male , Methimazole/administration & dosage , Mice , Mice, Inbred BALB C , Propylthiouracil/administration & dosage
16.
Pediatr Neonatol ; 60(4): 389-395, 2019 08.
Article En | MEDLINE | ID: mdl-30361144

BACKGROUND: This study investigated the prognostic parameters and beneficial effects of repeat plasma exchange in children with acute liver failure (ALF). METHODS: Twenty-three patients under 18 years of age admitted to National Taiwan University Hospital due to ALF from 2003 to 2016 were included in this retrospective analysis. RESULTS: Among the patients, 11 (48%) had native liver recovery (NLR), 9 (39.1%) died without liver transplant, and 3 (12.9%) received liver transplantation. The NLR group showed a lower proportion of idiopathic cases, lower peak ammonia level, higher peak alpha fetoprotein (AFP) level, and they had plasma exchange fewer times than the other groups. Receiver operating characteristic curve analyses yielded optimal cutoff values of plasma exchange (≤6 times), peak ammonia level (<190 µmol/L), and peak AFP level for predicting NLR in children with ALF. CONCLUSION: Pediatric ALF with idiopathic etiology, high peak ammonia level, and low peak AFP level are associated with fewer cases of NLR. Plasma exchange for more than six times probably offers little benefit with regard to patient survival if liver transplantation is not performed promptly.


Liver Failure, Acute/therapy , Liver Transplantation , Plasma Exchange , Adolescent , Ammonia/metabolism , Antithyroid Agents/toxicity , Chemical and Drug Induced Liver Injury/complications , Child , Child, Preschool , Female , Hemochromatosis/complications , Hepatitis, Viral, Human/complications , Hepatolenticular Degeneration/complications , Humans , Infant , Infant, Newborn , Liver Failure, Acute/etiology , Liver Failure, Acute/metabolism , Lymphohistiocytosis, Hemophagocytic/complications , Male , Metabolism, Inborn Errors/complications , Mortality , Prognosis , Propylthiouracil/toxicity , ROC Curve , Recovery of Function , Retrospective Studies , Taiwan , alpha-Fetoproteins/metabolism
17.
Pak J Pharm Sci ; 31(6): 2373-2377, 2018 Nov.
Article En | MEDLINE | ID: mdl-30473507

Methimazole (MMI) is a widely used drug for hyperthyroidism. However, its clinical use is associated with hepatotoxicity. Though the precise mechanism of hepatic damage is still far from clear, role of metabolic activation and reactive metabolites have been implicated. The present study was designed to investigate the role of enzyme induction in bioactivation based hepatotoxicity of methimazole in mice. Thirty male mice were randomly divided into five groups. Hepatotoxicity was induced by single intraperitoneal injection of methimazole (1000mg/kg). Pretreatment with rifampicin which is a potent enzyme inducer was carried out for 6 days prior to administration of methimazole. The extent of hepatic damage was determined by measuring serum alanine aminotransferase (ALT) and alkaline phosphatase (ALP) along with histopathological grading of liver samples. The elevated levels of biochemical markers by methimazole were potentiated by pretreatment with rifampicin. This potentiation of hepatic injury was also observed in liver histopathological examination. These findings suggest induction of microsomal enzymes as a potentiating factor of methimazole induced hepatotoxicity.


Antibiotics, Antitubercular/toxicity , Antithyroid Agents/toxicity , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Methimazole/toxicity , Rifampin/toxicity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/pathology , Drug Synergism , Liver/metabolism , Liver/pathology , Male , Mice, Inbred BALB C
18.
Toxicol Appl Pharmacol ; 358: 102-109, 2018 11 01.
Article En | MEDLINE | ID: mdl-30184474

Perchlorate pharmacology and toxicology studies date back at least 65 years in the peer-reviewed literature. Repeated studies in animals and humans have demonstrated perchlorate's mechanism of action, dose-response, and adverse effects over a range of doses. The first measurable effect of perchlorate is inhibition of iodine uptake to the thyroid gland. Adequate levels of thyroid hormones are critical for the development of the fetal nervous system. With sufficient dose and exposure duration, perchlorate can reduce thyroid hormones in the pregnant or non-pregnant woman via this mechanism. The developing fetus is the most sensitive life stage for chemical agents that affect iodide uptake to the thyroid. Perchlorate has a half-life of eight hours, is not metabolized, does not bioaccumulate, is not a mutagen or carcinogen, and is not reprotoxic or immunotoxic. More recently, epidemiological and biomonitoring studies have been published in the peer-reviewed literature characterizing the thyroidal effects of perchlorate and other goitrogens. While the results from most populations report no consistent association, a few studies report thyroidal effects at environmentally relevant levels of perchlorate. We reviewed the literature on health effects of perchlorate at environmental exposure levels, with a focus on exposures during pregnancy and neurodevelopmental effects. Based on the studies we reviewed, health effects are expected to only occur at doses substantially higher than environmental levels.


Antithyroid Agents/toxicity , Environmental Exposure/adverse effects , Health Status , Perchlorates/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Thyroid Gland/drug effects , Animals , Female , Humans , Pregnancy , Prenatal Exposure Delayed Effects/blood , Thyroid Gland/metabolism , Thyroid Hormones/blood
19.
J Appl Toxicol ; 38(12): 1529-1537, 2018 12.
Article En | MEDLINE | ID: mdl-30047161

Transcriptomics technologies have been used for risk assessment of chemicals, mainly to predict the modes of action (MOAs) of chemicals or identify biomarkers. Transcriptomics data may also be helpful to understand MOAs of chemicals at the molecular level in more detail. As an example of the known MOAs, there are two MOAs of thyroid toxicity: inhibition of thyroid hormone synthesis ("direct" effect) and hypermetabolism of thyroid hormone by enzyme induction in liver ("indirect" effect). In the present study, global profiles of gene expression were analyzed in rats treated with chemicals acting directly on the thyroid (thyroid peroxidase inhibitors such as propylthiouracil and methimazole) and chemicals acting indirectly on the thyroid (hepatic enzyme inducers such as phenobarbital and pregnenolone-16α-carbonitrile) using microarrays. Using a subtraction method between these two types of chemicals, we identified characteristic gene expression changes on the thyroid hormone synthesis pathway by direct-acting chemicals. Based on the functions of these genes, alterations of their expression seem to indicate the results of thyroid peroxidase inhibition, and might be helpful in more accurate evaluation of MOAs for thyroid toxicity.


Antithyroid Agents/toxicity , Liver/drug effects , Thyroid Gland/drug effects , Thyroid Hormones/biosynthesis , Transcriptome/drug effects , Animals , Enzyme Induction/drug effects , Gene Expression Profiling , Iodide Peroxidase/antagonists & inhibitors , Liver/enzymology , Male , Methimazole/toxicity , Microarray Analysis , Phenobarbital/toxicity , Propylthiouracil/toxicity , Rats, Wistar , Thyroid Gland/metabolism
20.
Toxicology ; 408: 70-79, 2018 09 01.
Article En | MEDLINE | ID: mdl-29990518

Anti-thyroid drugs (ATDs) therapy is necessary for pregnant women with hyperthyroidism. However, there is a lack of studies on developmental toxicity of ATDs. In this study, we observed the developmental toxicity of fetal liver induced by prenatal methimazole exposure (PME) in mice, and explored the potential mechanism. Pregnant Kunming mice were administered intragastrically with 4.5 or 18 mg/kg·d methimazole from gestational day (GD) 9∼18. After PME, the birth weights of the offspring mice were decreased, and the liver morphology, development indexes and metabolic function were all altered in different degree in the PME fetuses. Meanwhile, PME decreased the levels of serum and hepatic insulin-like growth factor 1 (IGF1), and reduced the gene expression of IGF1 downstream signaling pathway. Furthermore, the protein levels of phosphorylated-extracellular regulated protein kinases (p-ERK) and serine-threonine protein kinase (p-Akt) were also reduced. Furthermore, methimazole disturb hepatocyte differentiation, maturation and metabolic function through suppressing IGF1 signaling pathway in HepG2 cells. These results demonstrated that PME could induce fetal liver developmental toxicity, and the underlying mechanism was related to low-expression of hepatic IGF1 caused by methimazole, which mediated abnormal liver morphology and metabolic function.


Antithyroid Agents/toxicity , Insulin-Like Growth Factor I/metabolism , Liver/drug effects , Methimazole/toxicity , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gestational Age , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Insulin-Like Growth Factor I/genetics , Liver/embryology , Liver/metabolism , Maternal Exposure , Mice , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
...