Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters

Publication year range
1.
Toxicol Appl Pharmacol ; 491: 117064, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39122118

ABSTRACT

Propylthiouracil (PTU) and methimazole (MMI), two classical antithyroid agents possess risk of drug-induced liver injury (DILI) with unknown mechanism of action. This study aimed to examine and compare their hepatic toxicity using a quantitative system toxicology approach. The impact of PTU and MMI on hepatocyte survival, oxidative stress, mitochondrial function and bile acid transporters were assessed in vitro. The physiologically based pharmacokinetic (PBPK) models of PTU and MMI were constructed while their risk of DILI was calculated by DILIsym, a quantitative systems toxicology (QST) model by integrating the results from in vitro toxicological studies and PBPK models. The simulated DILI (ALT >2 × ULN) incidence for PTU (300 mg/d) was 21.2%, which was within the range observed in clinical practice. Moreover, a threshold dose of 200 mg/d was predicted with oxidative stress proposed as an important toxic mechanism. However, DILIsym predicted a 0% incidence of hepatoxicity caused by MMI (30 mg/d), suggesting that the toxicity of MMI was not mediated through mechanism incorporated into DILIsym. In conclusion, DILIsym appears to be a practical tool to unveil hepatoxicity mechanism and predict clinical risk of DILI.


Subject(s)
Antithyroid Agents , Chemical and Drug Induced Liver Injury , Hepatocytes , Methimazole , Oxidative Stress , Propylthiouracil , Propylthiouracil/toxicity , Propylthiouracil/pharmacokinetics , Methimazole/toxicity , Chemical and Drug Induced Liver Injury/etiology , Antithyroid Agents/toxicity , Humans , Hepatocytes/drug effects , Hepatocytes/metabolism , Oxidative Stress/drug effects , Models, Biological , Risk Assessment , Animals , Cell Survival/drug effects , Liver/drug effects , Liver/metabolism
2.
J Appl Toxicol ; 44(7): 1084-1103, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38563354

ABSTRACT

Although measurements of blood triiodothyronine (T3), thyroxine (T4), and thyroid-stimulating hormone (TSH) levels in rodent toxicity studies are useful for detection of antithyroid substances, assays for these measurements are expensive and can show high variability depending on blood sampling conditions. To develop more efficient methods for detecting thyroid disruptors, we compared histopathological and immunohistochemical findings in the thyroid and pituitary glands with blood hormone levels. Six-week-old male and female Sprague-Dawley rats (five rats per group) were treated with multiple doses of the thyroid peroxidase inhibitors propylthiouracil (PTU) and methimazole by gavage for 28 days. Significant decreases in serum T3 and T4 and increases in TSH were observed in the ≥1 mg/kg PTU and ≥3 mg/kg methimazole groups. An increase in TSH was also detected in male rats in the 0.3 mg/kg PTU group. Histopathological and immunohistochemical analyses revealed that follicular cell hypertrophy and decreased T4 and T3 expressions in the thyroid gland were induced at doses lower than doses at which significant changes in serum hormone levels were observed, suggesting that these findings may be more sensitive than blood hormone levels. Significant increases in thyroid weights, Ki67-positive thyroid follicular cell counts, and TSH-positive areas in the pituitary gland were detected at doses comparable with those at which changes in serum T4 and TSH levels were observed, indicating that these parameters may also be useful for evaluation of antithyroid effects. Combining these parameters may be effective for detecting antithyroid substances without relying on hormone measurements.


Subject(s)
Antithyroid Agents , Immunohistochemistry , Methimazole , Pituitary Gland , Propylthiouracil , Rats, Sprague-Dawley , Thyroid Gland , Thyrotropin , Thyroxine , Animals , Male , Antithyroid Agents/toxicity , Female , Thyroid Gland/drug effects , Thyroid Gland/pathology , Propylthiouracil/toxicity , Rats , Methimazole/toxicity , Thyrotropin/blood , Thyroxine/blood , Pituitary Gland/drug effects , Pituitary Gland/pathology , Iodide Peroxidase/antagonists & inhibitors , Triiodothyronine/blood , Thyroid Hormones/blood , Dose-Response Relationship, Drug
3.
Small ; 19(25): e2300236, 2023 06.
Article in English | MEDLINE | ID: mdl-36932895

ABSTRACT

The disruption of thyroid hormones because of chemical exposure is a significant societal problem. Chemical evaluations of environmental and human health risks are conventionally based on animal experiments. However, owing to recent breakthroughs in biotechnology, the potential toxicity of chemicals can now be evaluated using 3D cell cultures. In this study, the interactive effects of thyroid-friendly soft (TS) microspheres on thyroid cell aggregates are elucidated and their potential as a reliable toxicity assessment tool is evaluated. Using state-of-the-art characterization methods coupled with cell-based analysis and quadrupole time-of-flight mass spectrometry, it is shown that TS-microsphere-integrated thyroid cell aggregates exhibit improved thyroid function. Specifically, the responses of zebrafish embryos, which are used for thyroid toxicity analysis, and the TS-microsphere-integrated cell aggregates to methimazole (MMI), a known thyroid inhibitor, are compared. The results show that the thyroid hormone disruption response of the TS-microsphere-integrated thyroid cell aggregates to MMI is more sensitive compared with those of the zebrafish embryos and conventionally formed cell aggregates. This proof-of-concept approach can be used to control cellular function in the desired direction and hence evaluate thyroid function. Thus, the proposed TS-microsphere-integrated cell aggregates may yield new fundamental insights for advancing in vitro cell-based research.


Subject(s)
Thyroid Gland , Zebrafish , Animals , Humans , Antithyroid Agents/pharmacology , Thyroid Hormones/pharmacology , Methimazole/toxicity
4.
Drug Chem Toxicol ; 42(6): 615-623, 2019 Nov.
Article in English | MEDLINE | ID: mdl-29722569

ABSTRACT

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.


Subject(s)
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
5.
Turk J Med Sci ; 49(3): 715-722, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31023005

ABSTRACT

Background/aim: The aim of this study was to assess the effect of a combination use of methimazole (MMI) and selenium (Se) in the treatment of Graves' disease (GD). Materials and methods: A total of 103 newly diagnosed hyperthyroidism patients were randomized to MMI and MMI + Se combination groups. After treatment for 6 months, the levels of triiodothyronine (FT3), free thyroxine (FT4), thyrotropin receptor antibody (TRAb), thyroid peroxidase antibody (TPOAb), and thyroglobulin antibody (TGAb) were observed. An in vitro culture model of thyroid cells was established and the protein expression and mRNA levels of TRAb, TPOAb, and TGAb were determined by western blot and RT-PCR. Results: A significant decrease in the levels of FT3, FT4, TRAb, TPOAb, and TGAb were observed in both groups along with a marked increase in TSH levels. Furthermore, the in vitro experiments showed that the protein expression and mRNA levels of TRAb, TPOAb, and TGAb decreased significantly. Also, compared to the MMI group, there was a greater improvement of these indices in the MMI + Se group. Conclusion: We suggest that the combined use of MMI and Se could improve the thyroid activity in patients, which may provide an effective therapy for the treatment of GD in clinical settings.


Subject(s)
Antithyroid Agents/therapeutic use , Graves Disease/drug therapy , Methimazole/therapeutic use , Selenium/therapeutic use , Adult , Antithyroid Agents/administration & dosage , Autoantibodies/blood , Cell Survival/drug effects , Cells, Cultured , Dietary Supplements , Female , Graves Disease/pathology , Graves Disease/surgery , Humans , Male , Methimazole/administration & dosage , Methimazole/toxicity , Middle Aged , Pilot Projects , Receptors, Thyrotropin/immunology , Selenium/administration & dosage , Selenium/toxicity , Thyroid Gland/cytology , Thyroid Gland/pathology , Thyroid Gland/surgery , Thyroid Hormones/blood
6.
J Cell Biochem ; 119(7): 6249-6257, 2018 07.
Article in English | MEDLINE | ID: mdl-29663535

ABSTRACT

The purinergic system has an important role in the regulation of vascular functions. The interference of thyroid hormones in this system and in cardiovascular events has been studied in recent years. However, the mechanisms involved in vascular, purinergic, and oxidative changes in thyroid disorders are not completely understood. Therefore, the present study aimed to assess purinergic enzyme activity in platelets from rats with hypothyroidism and hyperthyroidism induced, respectively, by continuous exposure to methimazole (MMI) at 20 mg/100 mL or L-thyroxine at 1.2 mg/100 mL in drinking water for 1 month. Results showed that rats exposed to L-thyroxine had a significant decrease in NTPDase activity, wherein ATP hydrolysis was 53% lower and ADP hydrolysis was 40% lower. Moreover, ecto-5'-nucleotidase activity was decreased in both groups, by 39% in the hypothyroidism group and by 52% in the hyperthyroidism group. On the other hand, adenosine deaminase (ADA) activity was increased in hyperthyroidism (75%), and nucleotide pyrophosphatase/phosphodiesterase (NPP) activity was increased in animals with hypothyroidism (127%) and those with hyperthyroidism (128%). Our findings suggest that changes in purinergic enzyme and purine levels could contribute to the undesirable effects of thyroid disturbances. Moreover, oxidative stress and, in particular, a high level of ROS production, showed a causal relation with changes in ectonucleotidase activity and nucleotide and nucleoside levels.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine Deaminase/metabolism , Antigens, CD/metabolism , Apyrase/metabolism , Blood Platelets/enzymology , Hyperthyroidism/enzymology , Hypothyroidism/enzymology , Nucleotides/metabolism , Adenosine Triphosphate/metabolism , Animals , Hydrolysis , Hyperthyroidism/blood , Hyperthyroidism/chemically induced , Hypothyroidism/blood , Hypothyroidism/chemically induced , Male , Methimazole/toxicity , Oxidative Stress , Rats , Rats, Wistar
7.
Cereb Cortex ; 27(5): 3015-3027, 2017 05 01.
Article in English | MEDLINE | ID: mdl-27235101

ABSTRACT

Insufficient or excessive thyroid hormone (TH) levels during fetal development can cause long-term neurological and cognitive problems. Studies in animal models of perinatal hypo- and hyperthyroidism suggest that these problems may be a consequence of the formation of maladaptive circuitry in the cerebral cortex, which can persist into adulthood. Here we used mouse models of maternal hypo- and hyperthyroidism to investigate the long-term effects of altering thyroxine (T4) levels during pregnancy (corresponding to embryonic days 6.5-18.5) on thalamocortical (TC) axon dynamics in adult offspring. Because perinatal hypothyroidism has been linked to visual processing deficits in humans, we performed chronic two-photon imaging of TC axons and boutons in primary visual cortex (V1). We found that a decrease or increase in maternal serum T4 levels was associated with atypical steady-state dynamics of TC axons and boutons in V1 of adult offspring. Hypothyroid offspring exhibited axonal branch and bouton dynamics indicative of an abnormal increase in TC connectivity, whereas changes in hyperthyroid offspring were indicative of an abnormal decrease in TC connectivity. Collectively, our data suggest that alterations to prenatal T4 levels can cause long-term synaptic instability in TC circuits, which could impair early stages of visual processing.


Subject(s)
Hyperthyroidism/pathology , Hypothyroidism/pathology , Prenatal Exposure Delayed Effects/physiopathology , Synapses/physiology , Thalamus/pathology , Visual Cortex/pathology , Adult , Animals , Animals, Newborn , Antithyroid Agents/toxicity , Brain Mapping , Disease Models, Animal , Female , Gestational Age , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hyperthyroidism/diagnostic imaging , Hypothyroidism/diagnostic imaging , Image Processing, Computer-Assisted , Male , Methimazole/toxicity , Mice , Mice, Inbred C57BL , Neuroimaging , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/diagnostic imaging , Synapsins/genetics , Synapsins/metabolism , Thalamus/diagnostic imaging , Thyroxine/toxicity , Time Factors , Transduction, Genetic , Visual Cortex/diagnostic imaging
8.
Pharmacology ; 101(5-6): 309-321, 2018.
Article in English | MEDLINE | ID: mdl-29597200

ABSTRACT

Hypothyroidism is a common disorder that is associated with psychological disturbances such as dementia, depression, and psychomotor disorders. We recently found that chronic treatment with the T-type calcium channel enhancer SAK3 prevents the cholinergic neurodegeneration induced by a single intraperitoneal (i.p.) injection of methimazole (MMI; 75 mg/kg), thereby improving cognition. Here, we evaluated the acute effect of SAK3 on cognitive impairments and its mechanism of action following the induction of hypothyroidism. Hypothyroidism was induced by 2 injections of MMI (75 mg/kg, i.p.) administered once per week. Four weeks after the final MMI treatment, MMI-treated mice showed reduced serum thyroxine (T4) levels and cognitive impairments without depression-like behaviors. Although acute SAK3 (1.0 mg/kg, p.o.) administration failed to ameliorate the decreased T4 levels and histochemical destruction of the glomerular structure, acute SAK3 (1.0 mg/kg, p.o.) administration significantly reduced cognitive impairments in MMI-treated mice. Importantly, the α7 nicotinic acetylcholine receptor (nAChR)-selective inhibitor methyllycaconitine (MLA; 12 mg/kg, i.p.) and T-type calcium channel-specific blocker NNC 55-0396 (25 mg/kg, i.p.) antagonized the acute effect of SAK3 on memory deficits in MMI-treated mice. We also confirmed that acute SAK3 administration does not rescue reduced olfactory marker protein or choline acetyltransferase immunoreactivity levels in the olfactory bulb or medial septum. Taken together, these results suggest that SAK3 has the ability to improve the cognitive decline caused by hypothyroidism directly through activation of nAChR signaling and T-type calcium channels.


Subject(s)
Cognitive Dysfunction/prevention & control , Hypothyroidism/complications , Imidazoles/pharmacology , Methimazole/toxicity , Spiro Compounds/pharmacology , Acetylcholine/metabolism , Animals , Antithyroid Agents/toxicity , Benzimidazoles/pharmacology , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/metabolism , Cognitive Dysfunction/etiology , Cyclopropanes/pharmacology , Female , Hypothyroidism/chemically induced , Mice , Naphthalenes/pharmacology , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Signal Transduction/drug effects
9.
J Appl Toxicol ; 38(12): 1529-1537, 2018 12.
Article in English | MEDLINE | ID: mdl-30047161

ABSTRACT

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.


Subject(s)
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
10.
Pak J Pharm Sci ; 31(6): 2373-2377, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30473507

ABSTRACT

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.


Subject(s)
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
11.
J Neurosci ; 35(6): 2657-73, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673857

ABSTRACT

Although it is well known that injury induces the generation of a substantial number of new olfactory sensory neurons (OSNs) in the adult olfactory epithelium (OE), it is not well understood whether olfactory sensory input influences the survival and maturation of these injury-induced OSNs in adults. Here, we investigated whether olfactory sensory deprivation affected the dynamic incorporation of newly generated OSNs 3, 7, 14, and 28 d after injury in adult mice. Mice were unilaterally deprived of olfactory sensory input by inserting a silicone tube into their nostrils. Methimazole, an olfactotoxic drug, was also injected intraperitoneally to bilaterally ablate OSNs. The OE was restored to its preinjury condition with new OSNs by day 28. No significant differences in the numbers of olfactory marker protein-positive mature OSNs or apoptotic OSNs were observed between the deprived and nondeprived sides 0-7 d after injury. However, between days 7 and 28, the sensory-deprived side showed markedly fewer OSNs and mature OSNs, but more apoptotic OSNs, than the nondeprived side. Intrinsic functional imaging of the dorsal surface of the olfactory bulb at day 28 revealed that responses to odor stimulation were weaker in the deprived side compared with those in the nondeprived side. Furthermore, prevention of cell death in new neurons 7-14 d after injury promoted the recovery of the OE. These results indicate that, in the adult OE, sensory deprivation disrupts compensatory OSN regeneration after injury and that newly generated OSNs have a critical time window for sensory-input-dependent survival 7-14 d after injury.


Subject(s)
Homeostasis/physiology , Nerve Regeneration/physiology , Olfactory Receptor Neurons/injuries , Olfactory Receptor Neurons/physiology , Sensory Deprivation/physiology , Animals , Antithyroid Agents/toxicity , Apoptosis/physiology , Caspase Inhibitors/pharmacology , Cell Proliferation , Cell Survival , Male , Methimazole/toxicity , Mice , Mice, Inbred C57BL , Olfactory Mucosa/drug effects
12.
Regul Toxicol Pharmacol ; 80: 283-90, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27350053

ABSTRACT

The potencies of resorcinol, 6-propylthiouracil (PTU) and methimazole (MMI) for inducing developmental toxicity and neurotoxicity were compared in pregnant rats, regarded as valid model for human thyroid toxicity. Profound differences on maternal thyroid hormone levels (THs), maternal toxicity as well as developmental and neurotoxicity sequelae occurred. Resorcinol affected none of those end points. PTU and MMI caused significant effects. Therapy with either PTU or MMI during the first trimester of human pregnancy can cause reductions of maternal THs, accompanied by disruptions of prenatal development. Clinical MMI studies show sporadic evidence of teratogenic effects, with equivocal relation to thyroid peroxidase (TPO) inhibition. In recent decades no MMI associated prenatal toxicity has been reported, an outcome possibly related to carefully managed therapy. Orally administered resorcinol was rapidly absorbed, metabolized and excreted and was undetectable in the thyroid. In contrast, PTU or MMI accumulated. Resorcinol's potency to inhibit TPO was profoundly lower than that of PTU or MMI. Quantum chemical calculations may explain low resorcinol reactivity with TPO. Thus, distinctions in the target organ and the TPO inhibitory potency between these chemicals are likely contributing to different reductions of maternal THs levels and affecting the potency to cause developmental toxicity and neurotoxicity.


Subject(s)
Enzyme Inhibitors/toxicity , Iodide Peroxidase/antagonists & inhibitors , Iron-Binding Proteins/antagonists & inhibitors , Methimazole/toxicity , Propylthiouracil/toxicity , Resorcinols/toxicity , Thyroid Gland/drug effects , Abnormalities, Drug-Induced/etiology , Administration, Oral , Animals , Autoantigens/metabolism , Biomarkers/blood , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/metabolism , Female , Gestational Age , Humans , Iodide Peroxidase/metabolism , Iron-Binding Proteins/metabolism , Methimazole/administration & dosage , Methimazole/metabolism , Neurotoxicity Syndromes/etiology , Pregnancy , Propylthiouracil/administration & dosage , Propylthiouracil/metabolism , Rats , Resorcinols/administration & dosage , Resorcinols/metabolism , Risk Assessment , Thyroid Gland/enzymology , Thyroid Hormones/blood
13.
J Appl Toxicol ; 36(5): 702-15, 2016 May.
Article in English | MEDLINE | ID: mdl-26177832

ABSTRACT

Methimazole (MTZ), an anti-thyroid drug, is known to cause liver injury in humans. It has been demonstrated that MTZ-induced liver injury in Balb/c mice is accompanied by T helper (Th) 2 cytokine-mediated immune responses; however, there is little evidence for immune responses associated with MTZ-induced liver injury in rats. To investigate species differences in MTZ-induced liver injury, we administered MTZ with a glutathione biosynthesis inhibitor, L-buthionine-S,R-sulfoximine (BSO), to F344 rats and subsequently observed an increase in plasma alanine aminotransferase (ALT) and high-mobility group box 1 (HMGB1), which are associated with hepatic lesions. The hepatic mRNA expression of innate immune-related genes significantly increased in BSO- and MTZ-treated rats, but the change in Th2-related genes was not much greater than the change observed in the previous mouse study. Moreover, an increase in Kupffer cells and an induction of the phosphorylation of extracellular signal-regulated kinase (ERK)/c-Jun N-terminal kinase (JNK) proteins were accompanied by an increase in Toll-like receptor 4 (TLR4) expression, indicating that Kupffer cell activation occurs through HMGB1-TLR4 signaling. To elucidate the mechanism of liver injury in rats, gadolinium chloride, which inactivates the function of Kupffer cells, was administered before BSO and MTZ administration. The gadolinium chloride treatment significantly suppressed the increased ALT, which was accompanied by decreased hepatic mRNA expression related to innate immune responses and ERK/JNK phosphorylation. In conclusion, Kupffer cell-mediated immune responses are crucial factors for the exacerbation of MTZ-induced liver injury in rats, indicating apparent species differences in the immune-mediated exacerbation of liver injury between mice and rats.


Subject(s)
Antithyroid Agents/toxicity , Chemical and Drug Induced Liver Injury, Chronic/physiopathology , Kupffer Cells/drug effects , Liver/drug effects , Methimazole/toxicity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bilirubin/blood , Buthionine Sulfoximine , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gadolinium/pharmacology , HMGB1 Protein/blood , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Kupffer Cells/cytology , Male , Oxidative Stress/drug effects , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Wistar , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
14.
J Biochem Mol Toxicol ; 29(4): 173-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25545158

ABSTRACT

Liver injury is a deleterious adverse effect associated with methimazole administration, and reactive intermediates are suspected to be involved in this complication. Glyoxal is an expected reactive intermediate produced during methimazole metabolism. Current investigation was undertaken to evaluate the role of carnosine, metformin, and N-acetyl cysteine as putative glyoxal (carbonyl) traps, against methimazole-induced hepatotoxicity. Methimazole (100 mg/kg, intraperitoneally) was administered to intact and/or glutathione (GSH)-depleted mice and the role of glyoxal trapping agents was investigated. Methimazole caused liver injury as revealed by an increase in serum alanine aminotransferase and aspartate aminotransferase. Moreover, lipid peroxidation and protein carbonylation occurred significantly in methimazole-treated animals' liver. Hepatic GSH reservoirs were decreased, and inflammatory cells infiltration was observed in liver histopathology. Methimazole-induced hepatotoxicity was severe in GSH-depleted mice and accompanied with interstitial hemorrhage and necrosis of the liver. Glyoxal trapping agents effectively diminished methimazole-induced liver injury both in intact and/or GSH-depleted animals.


Subject(s)
Acetylcysteine/therapeutic use , Carnosine/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Metformin/therapeutic use , Methimazole/toxicity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Male , Mice , Necrosis , Oxidative Stress/drug effects , Protective Agents/therapeutic use
15.
Gen Comp Endocrinol ; 217-218: 28-36, 2015.
Article in English | MEDLINE | ID: mdl-25985894

ABSTRACT

The present study was conducted to investigate the effects and mechanisms of hypothyroidism, induced by administration of 0.2% methimazole through the food, on lipid metabolism in the liver of juvenile yellow catfish Pelteobagrus fulvidraco. To this end, yellow catfish were fed diets containing either 0 or 2g methimazole per kg of diet for 8weeks, respectively. The results showed that fish fed diet containing methimazole had a significant reduction in growth performance, plasma THs levels and hepatic lipid content. Meanwhile, methimazole treatment inhibited the activities of lipogenic enzymes (6-phosphogluconate dehydrogenase, glucose 6-phosphate dehydrogenase, malic enzyme, isocitrate dehydrogenase and fatty acid synthase) and the mRNA levels of genes involved in lipogenesis (6-phosphogluconate dehydrogenase, glucose 6-phosphate dehydrogenase, fatty acid synthase, acetyl-CoA carboxylase α, sterol-regulator element-binding protein-1 and liver X receptor), but increased lipolytic enzyme (carnitine palmitoyltransferase 1) activity and the expression of genes involved in lipolysis (carnitine palmitoyltransferase 1a, hormone-sensitive lipase and peroxisome proliferators-activated receptor α). Thus, our study indicated that dietary methimazole-induced hypothyroidism could disturb the normal processes of lipid metabolism at the enzymatic and molecular levels in yellow catfish, and the reduced hepatic lipid content by hypothyroidism was attributable to the down-regulation of lipogenesis and up-regulation of lipolysis.


Subject(s)
Diet , Hypothyroidism/metabolism , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Lipolysis/drug effects , Liver/drug effects , Methimazole/toxicity , Adipogenesis/drug effects , Adipogenesis/physiology , Animals , Antithyroid Agents/toxicity , Catfishes/growth & development , Catfishes/metabolism , Down-Regulation , Hypothyroidism/chemically induced , Hypothyroidism/pathology , Liver/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
16.
Mol Pain ; 10: 38, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24943008

ABSTRACT

BACKGROUND: Thyroid hormones are essential for the maturation and functions of the central nervous system. Pain sensitivity is related to the thyroid status. However, information on how thyroid hormones affect pain processing and synaptic transmission in the anterior cingulate cortex (ACC) is limited. Nociceptive threshold and synaptic transmission in the ACC were detected in the experimental hypothyroidism (HT) mice. RESULTS: HT was induced by methimazole and potassium perchlorate in distilled drinking water for 4 weeks. The threshold of pain perception to hot insults, but not mechanical ones, decreased in hypothyroid mice. After treatment with tri-iodothyronine (T3) or thyroxine (T4) for 2 weeks, thermal pain threshold recovered. Electrophysiological recordings revealed enhanced glutamatergic synaptic transmission and reduced GABAergic synaptic transmission in the ACC. Supplementation with T3 or T4 significantly rescued this synaptic transmission imbalance. In the same model, HT caused the up-regulation of the GluR1 subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor and NR2B-containing N-methyl-D-aspartate receptors, but it down-regulated γ-aminobutyric acid A receptors in the ACC. Supplementation with T3 or T4 notably recovered the levels of above proteins. CONCLUSIONS: These results suggest that HT promotes hypersensitivity to noxious thermal, and that supplementation with T3 or T4 rescues the imbalance between excitatory and inhibitory transmission in the ACC.


Subject(s)
Gyrus Cinguli/physiopathology , Hypothyroidism/pathology , Pain Threshold/physiology , Synaptic Transmission/physiology , Animals , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gyrus Cinguli/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hypothyroidism/blood , Hypothyroidism/complications , Hypothyroidism/etiology , In Vitro Techniques , Male , Methimazole/toxicity , Mice , Mice, Inbred C57BL , Pain Threshold/drug effects , Perchlorates/toxicity , Potassium Compounds/toxicity , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/drug effects , Thyroxine/blood , Thyroxine/pharmacology , Triiodothyronine/blood , Triiodothyronine/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
17.
FASEB J ; 27(2): 684-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23150524

ABSTRACT

Antithyroid medications are the preferred therapy for the treatment of Graves' disease during pregnancy. Propylthiouracil (PTU) is favored over methimazole (MMI) due to potential teratogenic concerns with MMI. This study was to determine the teratogenic potential of MMI and PTU using a validated Xenopus tropicalis embryo model. Embryos were exposed to 1 mM PTU (EC(50)=0.88 mM), 1 mM MMI, or vehicle control (water) from stages 2 to 45. Treated embryos were examined for gross morphological defects, ciliary function, and gene expression by in situ hybridization. Exposure to PTU, but not MMI, led to cardiac and gut looping defects and shortening along the anterior-posterior axis. PTU exposure during gastrulation (stage 8-12.5) was identified as the critical period of exposure leading to left-right (LR) patterning defects. Abnormal cilia polarization, abnormal cilia-driven leftward flow at the gastrocoel roof plate (GRP), and aberrant expression of both Coco and Pitx2c were associated with abnormal LR symmetry observed following PTU exposure. PTU is teratogenic during late blastula, gastrulation, and neurulation; whereas MMI is not. PTU alters ciliary-driven flow and disrupts the normal genetic program involved in LR axis determination. These studies have important implications for women taking PTU during early pregnancy.


Subject(s)
Antithyroid Agents/toxicity , Body Patterning/drug effects , Propylthiouracil/toxicity , Teratogens/toxicity , Xenopus/embryology , Animals , Antithyroid Agents/administration & dosage , Body Patterning/genetics , Cilia/drug effects , Digestive System Abnormalities/chemically induced , Digestive System Abnormalities/embryology , Female , Gene Expression Regulation, Developmental/drug effects , Graves Disease/complications , Graves Disease/drug therapy , Heart Defects, Congenital/chemically induced , Heart Defects, Congenital/embryology , Humans , Methimazole/administration & dosage , Methimazole/toxicity , Models, Animal , Pregnancy , Pregnancy Complications/drug therapy , Propylthiouracil/administration & dosage , Time Factors , Triiodothyronine/pharmacology , Xenopus/genetics
18.
Toxicol Pathol ; 42(6): 970-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24067673

ABSTRACT

Thionamides such as propylthiouracil (PTU) and methimazole (MMI) have been used for more than 50 years to treat the more common causes of thyrotoxicosis/hyperthyroidism such as Graves' disease. Serious adverse effects associated with thionamides in humans include idiosyncratic liver damage, agranulocytosis, aplastic anemia, and vasculitis. Both prospective and retrospective clinical studies with these drugs have failed to identify predictive biomarker for these adverse effects. To assess whether rat is a good model for predicting drug-related adverse events in the liver and in the bone marrow, we conducted a comprehensive study in male rats with multiple doses of PTU and MMI. As expected, euthyroid animals became hypothyroid along with several secondary changes associated with hypothyroidism. There were slight reductions in red blood cell parameters along with some marginal effects on the bone marrow elements. However, there was no evidence of significant neutropenia and liver injury in both PTU-treated and MMI-treated cohorts. MMI-related effects were noted in the seminiferous tubules of the testes. Overall, 1-month daily treatment of euthyroid rats with PTU or MMI resulted in hypothyroidism, minor bone marrow effects, and several secondary effects associated with hypothyroidism, but without any evidence of adverse effects reported in humans including liver injury and agranulocytosis.


Subject(s)
Methimazole/toxicity , Propylthiouracil/toxicity , Testis/drug effects , Thyroid Gland/drug effects , Animals , Male , Methimazole/administration & dosage , Methimazole/blood , Methimazole/pharmacokinetics , Propylthiouracil/administration & dosage , Propylthiouracil/blood , Propylthiouracil/pharmacokinetics , Rats , Rats, Wistar , Testis/chemistry , Testis/pathology , Thyroid Gland/chemistry , Thyroid Gland/pathology , Toxicity Tests
19.
Bioorg Med Chem ; 22(9): 2809-15, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24703463

ABSTRACT

In this study, we synthesized 4 methimazole (2-mercapto-1-methylimidazole, MMI) derivatives. The kinetics of inhibition on mushroom tyrosinase by methimazole and its derivatives were investigated. The results indicated that tert-butyl 3-methyl-2-sulfanylidene-2,3-dihydro-1H-imidazole-1-carboxylate (compound 3; 3), 2-mercaptoimidazole (MI; compound 1; 1) and MMI (compound 2; 2) significantly inhibited tyrosinase activity in a dose-dependent manner, exhibiting an IC50 value of 1.50mM, 4.11 mM, and 1.43 mM. However, compound 4 (4), compound 5 (5), and compound 6 (6) exerted no inhibitory effect on mushroom tyrosinase activity. Kinetic analysis indicated that 3 was a noncompetitive tyrosinase inhibitor, whereas both 1 and 2 were exhibited as mixed-type tyrosinase inhibitors. Furthermore, 3 exerted a potent inhibitory effect on intracellular melanin formation in the B16/F10 murine melanoma cells and did not cause cytotoxicity, as 1 and 2 did.


Subject(s)
Enzyme Inhibitors/chemistry , Melanins/metabolism , Methimazole/chemistry , Monophenol Monooxygenase/antagonists & inhibitors , Agaricales , Animals , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/toxicity , Kinetics , Methimazole/chemical synthesis , Methimazole/toxicity , Mice , Monophenol Monooxygenase/metabolism
20.
Birth Defects Res B Dev Reprod Toxicol ; 101(4): 300-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24980470

ABSTRACT

BACKGROUND: Propylthiouracil (PTU) and methimazole (MMI) are antithyroid drugs used to treat hyperthyroidism. Despite the widespread use of PTU and MMI during pregnancy, modest clinical data and less animal data are available on the teratogenic potential of these drugs. METHODS: We evaluated the teratogenicity of in utero exposure to PTU or MMI in mice and rats. First, pregnant C57Bl/6 mice were treated daily with PTU (10 or 100 mg/kg), MMI (2 or 20 mg/kg), or vehicle from gestation day (GD) 6 to 16. GD 18 fetuses were evaluated for gross and histopathological abnormalities. Next, pregnant Sprague-Dawley rats were treated daily with PTU (50 or 100 mg/kg), MMI (10 or 20 mg/kg), or vehicle from GD 6 to 19, followed by evaluation for gross and histopathological abnormalities at GD 20. RESULTS: In mice treated with PTU or MMI, no significant histopathological abnormalities or external gross malformations, and no adverse effects on placental weight, litter size, resorption rates, or fetal weight were observed at GD 18. In rats, no adverse effects on litter size, placental weights, or maternal body weights were observed with either PTU or MMI treatment. PTU treatment (50 and 100 mg/kg) and MMI (10 mg/kg) treatment resulted in a decrease in crown-rump length in rat fetuses but no external gross malformations or histopathological abnormalities were observed. CONCLUSION: We did not observe either gross external malformations or histopathological malformations in mice or rats treated long-term with high doses of PTU or MMI during pregnancy.


Subject(s)
Antithyroid Agents/toxicity , Hyperthyroidism/drug therapy , Methimazole/toxicity , Pregnancy Complications/chemically induced , Propylthiouracil/toxicity , Animals , Antithyroid Agents/pharmacology , Female , Methimazole/pharmacology , Mice , Mice, Inbred C57BL , Pregnancy , Propylthiouracil/pharmacology , Rats , Rats, Sprague-Dawley , Teratogens/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL