Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
J Appl Toxicol ; 44(8): 1214-1235, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38654465

ABSTRACT

Thiazolidinediones (TZDs) (e.g. pioglitazone and rosiglitazone), known insulin sensitiser agents for type II diabetes mellitus, exhibit controversial effects on cardiac tissue. Despite consensus on their association with increased heart failure risk, limiting TZD use in diabetes management, the underlying mechanisms remain uncharacterised. Herein, we report a comprehensive in vitro investigation utilising a novel toxicoproteomics pipeline coupled with cytotoxicity assays in human adult cardiomyocytes to elucidate mechanistic insights into TZD cardiotoxicity. The cytotoxicity assay findings showed a significant loss of mitochondrial adenosine triphosphate production upon exposure to either TZD agents, which may underpin TZD cardiotoxicity. Our toxicoproteomics analysis revealed that mitochondrial dysfunction primarily stems from oxidative phosphorylation impairment, with distinct signalling mechanisms observed for both agents. The type of cell death differed strikingly between the two agents, with rosiglitazone exhibiting features of caspase-dependent apoptosis and pioglitazone implicating mitochondrial-mediated necroptosis, as evidenced by the protein upregulation in the phosphoglycerate mutase family 5-dynamin-related protein 1 axis. Furthermore, our analysis revealed additional mechanistic aspects of cardiotoxicity, showcasing drug specificity. The downregulation of various proteins involved in protein machinery and protein processing in the endoplasmic reticulum was observed in rosiglitazone-treated cells, implicating proteostasis in the rosiglitazone cardiotoxicity. Regarding pioglitazone, the findings suggested the potential activation of the interplay between the complement and coagulation systems and the disruption of the cytoskeletal architecture, which was primarily mediated through the integrin-signalling pathways responsible for pioglitazone-induced myocardial contractile failure. Collectively, this study unlocks substantial mechanistic insight into TZD cardiotoxicity, providing the rationale for future optimisation of antidiabetic therapies.


Subject(s)
Cardiotoxicity , Myocytes, Cardiac , Pioglitazone , Proteomics , Rosiglitazone , Thiazolidinediones , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Thiazolidinediones/toxicity , Proteomics/methods , Rosiglitazone/pharmacology , Hypoglycemic Agents/toxicity , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Mitochondria/drug effects , Mitochondria/metabolism
2.
Metabolomics ; 20(2): 24, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38393619

ABSTRACT

INTRODUCTION: Thiazolidinediones (TZDs), represented by pioglitazone and rosiglitazone, are a class of cost-effective oral antidiabetic agents posing a marginal hypoglycaemia risk. Nevertheless, observations of heart failure have hindered the clinical use of both therapies. OBJECTIVE: Since the mechanism of TZD-induced heart failure remains largely uncharacterised, this study aimed to explore the as-yet-unidentified mechanisms underpinning TZD cardiotoxicity using a toxicometabolomics approach. METHODS: The present investigation included an untargeted liquid chromatography-mass spectrometry-based toxicometabolomics pipeline, followed by multivariate statistics and pathway analyses to elucidate the mechanism(s)of TZD-induced cardiotoxicity using AC16 human cardiomyocytes as a model, and to identify the prognostic features associated with such effects. RESULTS: Acute administration of either TZD agent resulted in a significant modulation in carnitine content, reflecting potential disruption of the mitochondrial carnitine shuttle. Furthermore, perturbations were noted in purine metabolism and amino acid fingerprints, strongly conveying aberrations in cardiac energetics associated with TZD usage. Analysis of our findings also highlighted alterations in polyamine (spermine and spermidine) and amino acid (L-tyrosine and valine) metabolism, known modulators of cardiac hypertrophy, suggesting a potential link to TZD cardiotoxicity that necessitates further research. In addition, this comprehensive study identified two groupings - (i) valine and creatine, and (ii) L-tryptophan and L-methionine - that were significantly enriched in the above-mentioned mechanisms, emerging as potential fingerprint biomarkers for pioglitazone and rosiglitazone cardiotoxicity, respectively. CONCLUSION: These findings demonstrate the utility of toxicometabolomics in elaborating on mechanisms of drug toxicity and identifying potential biomarkers, thus encouraging its application in the toxicological sciences. (245 words).


Subject(s)
Diabetes Mellitus, Type 2 , Heart Failure , Thiazolidinediones , Humans , Rosiglitazone/therapeutic use , Pioglitazone , Myocytes, Cardiac , Cardiotoxicity/complications , Cardiotoxicity/drug therapy , Diabetes Mellitus, Type 2/complications , Metabolomics , Thiazolidinediones/toxicity , Heart Failure/chemically induced , Amino Acids , Biomarkers , Carnitine , Valine
3.
Neurotoxicology ; 93: 211-221, 2022 12.
Article in English | MEDLINE | ID: mdl-36228751

ABSTRACT

INTRODUCTION: Chemotherapy-induced peripheral neuropathy (CIPN) is a common side effect of anticancer drugs which affect the peripheral nervous system, as occurs with cisplatin treatment. Nowadays, one strategy in development to prevent, minimize and/or revert CIPN is neuroprotection. Therefore, we have evaluated the signaling pathways involved in CIPN and the effect of rosiglitazone, a peroxisome proliferator-activated receptor γ (PPAR-γ) agonist. METHODS: Dorsal Root Ganglia (DRG) were harvested from Wistar rats (Rattus norvegicus), the cells were dissociated, plated, and maintained with nerve growth factor for 9 days. On day 8, the cells were treated with cisplatin, rosiglitazone and/or T0070907 (PPAR-γ antagonist) for 24 h. The cell viability was measured by trypan blue exclusion method, the mRNA was quantified by real-time RT-PCRq and the release of TNF-α and calcitonin gene-related peptide (CGRP) was evaluated by ELISA. RESULTS: Cisplatin, rosiglitazone or T0070907 treatments did not decreased the cell viability on the primary DRG cultures cells. Cisplatin treatment induced a decrease of PPAR-γ and -ß/δ mRNA, while the co-treatment with rosiglitazone inhibited this cisplatin-induced effect. Moreover, T0070907 did not change the observed results, indicating that the rosiglitazone's effect could be due to mechanisms beyond PPAR-γ activation. Also, the rosiglitazone effect is not exclusively to DRG cells since there was an increase of PPAR-γ mRNA expression in 3T3-L1 cells. Furthermore, rosiglitazone did not modulate the cisplatin decrease neuronal function of DRG cells (TNF-α and CGRP release). CONCLUSION: Cisplatin decreased the gene expression of PPAR-γ and -ß/δ, while the rosiglitazone treatment inhibited these effects via PPAR-γ independent pathway. Rosiglitazone did not show improvement in modulation of TNF-α or CGRP release impaired by cisplatin.


Subject(s)
Neurotoxicity Syndromes , Thiazolidinediones , Rats , Animals , Rosiglitazone/pharmacology , Calcitonin Gene-Related Peptide , Tumor Necrosis Factor-alpha/metabolism , Ganglia, Spinal , Thiazolidinediones/toxicity , Thiazolidinediones/therapeutic use , Cisplatin/toxicity , Rats, Wistar , PPAR gamma/genetics , Hypoglycemic Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , RNA, Messenger
4.
Fundam Clin Pharmacol ; 36(1): 143-149, 2022 Feb.
Article in English | MEDLINE | ID: mdl-33969534

ABSTRACT

Thiazolidinediones are well-known anti-diabetic drugs. However, they are not widely used due to their cardiotoxic effects. Therefore, in this study, we aimed to determine the molecular toxicological alterations induced in the mouse hearts after thiazolidinedione administration. Balb/c mice received doses clinically equivalent to those given to humans of the most commonly used thiazolidinediones, pioglitazone, and rosiglitazone for 30 days. After that, RNA samples were isolated from the hearts. The mRNA expression of cytochrome (cyp) p450 genes that synthesize the cardiotoxic 20-hydroxyeicosatetraenoic acid (20-HETE) in addition to 92 cardiotoxicity biomarker genes were analyzed using quantitative polymerase chain reaction array technique. The analysis demonstrated that thiazolidinediones caused a significant upregulation (p < 0.5) of the mRNA expression of cyp1a1, cyp4a12, itpr1, ccl7, ccr1, and b2 m genes. In addition, thiazolidinediones caused a significant (p < 0.05) downregulation of the mRNA expression of adra2a, bsn, col15a1, fosl1, Il6, bpifa1, plau, and reg3b genes. The most affected gene was itpr1 gene, which was upregulated by pioglitazone and rosiglitazone by sevenfold and 3.5-fold, respectively. In addition, pioglitazone caused significant upregulation of (p < 0.05) hamp, ppbp, psma2, sik1, timp1, and ucp1 genes, which were not affected significantly (p > 0.05) by rosiglitazone administration. In conclusion, this study showed that thiazolidinediones induce toxicological molecular alterations in the mouse hearts, such as the induction of cyp450s that synthesize 20-HETE, chemokine activation, inflammatory responses, blood clotting, and oxidative stress. These findings may help us understand the mechanism of cardiotoxicity involved in thiazolidinedione administration.


Subject(s)
Pharmaceutical Preparations , Thiazolidinediones , Animals , Glycoproteins , Hypoglycemic Agents/toxicity , Mice , Phosphoproteins , Rosiglitazone/toxicity , Thiazolidinediones/toxicity
5.
Bioorg Med Chem Lett ; 30(23): 127561, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32961322

ABSTRACT

Thiazolidinedione (TZD) has been an interesting scaffold due to its proven antidiabetic activity and encouraging findings in anticancer drug discovery. We synthesised benzylidene thiazolidinedione derivatives which exhibited excellent antiproliferative effects in chronic myeloid leukemic cells K562 and the most active compounds 3t and 3x had GI50 value of 0.9 and 0.23 µM respectively. Both the compound was found to arrest the growth of K562 cells in G0/G1 phase in a time and dose dependent manner. Further, western blot analysis revealed that 3t and 3x could also inhibit the expression of cell proliferation markers, PCNA and Cyclin D1 and compound 3x up-regulated apoptosis markers, cleaved PARP1 and activated caspase 3, which could be a possible mechanism for the excellent antiproliferative effects exhibited by these compounds. In vitro combination studies of 3t and 3x with Imatinib found to potentiate the antitumor effects of Imatinib. Further in vivo efficacy in K562 xenografts, of 3t and 3x alone and in combination with Imatinib was found to be promising and far better than control group and combination treatment was found to be more effective as compared to only Imatinib treated or test compound treated animals. Thus, our findings suggest that these compounds are promising antitumor agents and could help to enhance the anticancer effects of Imatinib and other tyrosine kinase inhibitors, when used in combination.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzylidene Compounds/therapeutic use , Imatinib Mesylate/therapeutic use , Neoplasms/drug therapy , Thiazolidinediones/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Benzylidene Compounds/chemical synthesis , Benzylidene Compounds/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Mice, Nude , Molecular Structure , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/toxicity , Xenograft Model Antitumor Assays
6.
CNS Neurol Disord Drug Targets ; 18(10): 798-807, 2019.
Article in English | MEDLINE | ID: mdl-31742497

ABSTRACT

BACKGROUND: Epilepsy is a serious and common neurological disorder threatening the health of humans. Despite enormous progress in epileptic research, the anti-epileptic drugs present many limitations. These limitations prompted the development of more safer and effective AEDs. METHODS: A series of N-substituted (Z)-5-(benzo[d][1,3]dioxol-5-ylmethylene)- 2-thioxothiazolidin-4- one derivatives and 5-substituted-thioxothiazolidindione derivatives were designed, synthesized and tested for anticonvulsant activity against maximal electroshock (MES) and subcutaneous pentylenetetrazole (scPTZ). Neurotoxicity was determined by the rotarod test. RESULTS: Among them, the most potent 4e displayed high protection against MES-induced seizures with an ED50 value of 9.7 mg/kg and TD50 value of 263.3 mg/kg, which provided 4e with a high protective index (TD50/ED50) of 27.1 comparable to reference antiepileptic drugs. 4e clearly inhibits the NaV1.1 channel in vitro. The molecular docking study was conducted to exploit the results. CONCLUSION: Stiripentol is a good lead compound for further structural modification. Compound 4e was synthesized, which displayed remarkable anticonvulsant activities, and the NaV1.1 channel inhibition was involved in the mechanism of action of 4e.


Subject(s)
Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacology , Seizures/prevention & control , Thiazolidinediones/chemical synthesis , Thiazolidinediones/pharmacology , Animals , Anticonvulsants/toxicity , Dose-Response Relationship, Drug , Electroshock , Mice , Molecular Docking Simulation , NAV1.1 Voltage-Gated Sodium Channel/drug effects , Pentylenetetrazole , Seizures/chemically induced , Structure-Activity Relationship , Thiazolidinediones/toxicity
7.
Biomolecules ; 9(8)2019 07 27.
Article in English | MEDLINE | ID: mdl-31357635

ABSTRACT

Thiazolidinedione 49 (TD49), a newly synthesized algicide, shows strong toxicity at low concentrations of 0.1-2.0 µM. However, its potential effects on non-target species at the transcript level were not investigated. Differentially expressed genes (DEGs) in the gills of the bay scallop, Argopecten irradians, were accessed after treatment with 0.68 µM TD49 for up to 48 h. Following exposure, it was observed that 5214 genes were upregulated and 3497 were downregulated. Functional enrichment analysis revealed that the apoptosis pathway was activated. The extrinsic apoptosis pathway was activated and the survival factors related pathway was suppressed. Furthermore, gene expressions related to ATP-binding cassette, nuclear factor erythroid 2-related factor, B cell lymphoma-2 family protein, glutathione reductase, glutathione peroxidase, catalase, NADPH2:quinone reductase, and superoxide dismutase were decreased. Conversely, gene expressions related to FAS-associated death domain protein, glutathione S-transferase, caspase 6, 8, cytochrome P450 1A1, and 2C8 were increased. These results comprehensively demonstrated the toxicity of the novel algicide TD49, and should draw the attention of researchers to the importance of analyzing the potential impact of chemical compounds as algicides to control the proliferation of harmful algae, due to the secondary pollution caused by their application.


Subject(s)
Gene Expression Profiling/methods , Herbicides/toxicity , Pectinidae/genetics , Thiazolidinediones/toxicity , Animals , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Gills , High-Throughput Nucleotide Sequencing , Pectinidae/drug effects , Sequence Analysis, RNA
8.
J Appl Toxicol ; 39(9): 1348-1361, 2019 09.
Article in English | MEDLINE | ID: mdl-31231834

ABSTRACT

CJ-12,918, a 5-lipoxygenase (5-LO) inhibitor, caused cataracts during a 1-month safety assessment studies in rats whereas the structurally similar ZD-2138 was without effect. For CJ-12,918 analogs, blocking different sites of metabolic liability reduced (CJ-13,454) and eliminated (CJ-13,610) cataract formation in both rats and dogs. Using this chemical series as a test set, models and mechanisms of toxicity were first explored by testing the utility of ex vivo rat lens explant cultures as a safety screen. This model overpredicted the cataractogenic potential of ZD-2138 due to appreciably high lens drug levels and was abandoned in favor of a mechanism-based screen. Perturbations in lens sterol content, from a decline in lathosterol content, preceded cataract formation suggesting CJ-12,918 inhibited lens cholesterol biosynthesis (LCB). A 2-day bioassay in rats using ex vivo LCB assessments showed that the level of LCB inhibition was correlated with incidence of cataract formation in animal studies by these 5-LO inhibitors. Thereafter, this 2-day bioassay was applied to other pharmaceutical programs (neuronal nitric oxide synthase, sorbitol dehydrogenase inhibitor, squalene synthetase inhibitor and stearoyl-CoA desaturase-1 inhibitors/D4 antagonists) that demonstrated cataract formation in either rats or dogs. LCB inhibition >40% was associated with a high incidence of cataract formation in both rats and dogs that was species specific. Bioassay sensitivity/specificity were further explored with positive (RGH-6201/ciglitazone/U18666A) and negative (tamoxifen/naphthalene/galactose) mechanistic controls. This body of work over two decades shows that LCB inhibition was a common mechanism of cataract formation by pharmaceutical agents and defined a level of inhibition >40% that was typically associated with causing cataracts in safety assessment studies typically ≥1 month.


Subject(s)
Cataract/chemically induced , Cholesterol/biosynthesis , Cholesterol/toxicity , Enzyme Inhibitors/toxicity , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , Thiazolidinediones/toxicity , Animals , Animals, Laboratory , Cataract/metabolism , Dogs , Female , Male , Pharmaceutical Preparations , Rats , Rats, Sprague-Dawley
9.
J Toxicol Sci ; 43(5): 339-351, 2018.
Article in English | MEDLINE | ID: mdl-29743445

ABSTRACT

Troglitazone, a member of the thiazolidinedione class of antidiabetic drugs, was withdrawn from the market because it causes severe liver injury. One of the mechanisms for this adverse effect is thought to be mitochondrial toxicity. To investigate the characteristics of troglitazone-induced liver toxicity in more depth, the toxicological effects of troglitazone on hepatocytes and liver mitochondria were investigated using a rat model of type 2 diabetes mellitus (T2DM). Troglitazone was found to increase mitochondrial permeability transition (MPT) in the liver mitochondria of diabetic rats to a greater extent than in control rats, whereas mitochondrial membrane potential and oxidative phosphorylation were not affected. To identify the factors associated with this increase in susceptibility to MPT in diabetic rats, we assessed the oxidative status of the liver mitochondria and found a decrease in mitochondrial glutathione content and an increase in phospholipid peroxidation. Moreover, incorporation of oxidized cardiolipin, a mitochondrion-specific phospholipid, was involved in the troglitazone-induced alteration in susceptibility to MPT. In conclusion, liver mitochondria display disease-associated mitochondrial lipid peroxidation in T2DM, which facilitates the higher susceptibility to troglitazone-induced MPT. Thus, greater susceptibility of liver mitochondria may be a host factor leading to troglitazone-induced hepatotoxicity in T2DM.


Subject(s)
Chromans/toxicity , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/toxicity , Lipid Peroxidation , Mitochondria, Liver/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Thiazolidinediones/toxicity , Animals , Cardiolipins/metabolism , Chromans/adverse effects , Disease Models, Animal , Glutathione/metabolism , Hepatocytes/drug effects , Hypoglycemic Agents/adverse effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Permeability Transition Pore , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Phospholipids/metabolism , Rats, Zucker , Thiazolidinediones/adverse effects , Troglitazone
10.
Toxicol Appl Pharmacol ; 338: 204-213, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29146462

ABSTRACT

Drug-induced liver injury (DILI) has been the most frequent cause of post-marketing drug withdrawals in the last 50years. The multifactorial nature of events that precede severe liver injury in human patients is difficult to model in rodents due to a variety of confounding or contributing factors that include disease state, concurrent medications, and translational species differences. In retrospective analyses, a consistent risk factor for DILI has been the inhibition of the Bile Salt Export Pump (BSEP). One compound known for potent BSEP inhibition and severe DILI is troglitazone. The purpose of the current study is to determine if serum profiling of 19 individual bile acids by liquid chromatography-mass spectrometry (LC/MS) can detect perturbations in bile acid homeostasis in rats after acute intravenous (IV) administration of vehicle or 5, 25, or 50mg/kg troglitazone. Minimal serum transaminase elevations (approximately two-fold) were observed with no evidence of microscopic liver injury. However, marked changes in individual serum bile acids occurred, with dose-dependent increases in the majority of the bile acids profiled. When compared to predose baseline values, tauromuricholic acid and taurocholic acid had the most robust increase in serum levels and dynamic range, with a maximum fold increase from baseline of 34-fold and 29-fold, respectively. Peak bile acid increases occurred within 2hours (h) after dosing and returned to baseline values before 24h. In conclusion, serum bile acid profiling can potentially identify a mechanistic risk of clinical DILI that could be poorly detected by traditional toxicity endpoints.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 11/antagonists & inhibitors , Bile Acids and Salts/blood , Chemical and Drug Induced Liver Injury/etiology , Risk Assessment , Animals , Chromans/toxicity , Female , Male , Rats , Rats, Sprague-Dawley , Thiazolidinediones/toxicity , Troglitazone
11.
Hum Exp Toxicol ; 37(8): 827-835, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29025280

ABSTRACT

Rosiglitazone is in the thiazolidinedione class of drugs used in the treatment of type 2 diabetes mellitus. It works as an insulin sensitizer by binding to the peroxisome proliferator-activated receptor gamma. We investigated the effects of prenatally administered rosiglitazone on pyramidal cell numbers and morphologies in the hippocampus at postnatal period using histochemical and stereological techniques, congenital morphological properties and the number of offspring in rats. Eighteen female rats were grouped into control (C), low-dose rosiglitazone (LDR) and high-dose rosiglitazone (HDR). LDR pregnant rats received 2 mg/kg/day of rosiglitazone via oral gavage during the first 16 days of the pregnancy. HDR rats received 5 mg/kg/day. The infants were grouped into newborn (NB), 4 week (4 W) and 12 week (12 W). A side from histopathologic and congenital assessments, stereological analyses were performed using the optical fractionator method. Congenital anomaly was not detected in any of the rosiglitazone treatment groups, and their number of offspring was similar to that of the C group. Stereological counts revealed a significant reduction in the number of hippocampal pyramidal cells in the C and LDR groups but not in the HDR group until birth to 12th week. When NB groups were compared, the number of pyramidal cells in the HDRNB group was less than those in the LDRNB and CNB groups. HDR affected apoptosis or the proliferation and maturation of progenitor cells to the pyramidal neuron during neurodevelopment in the hippocampus, whereas LDR did not adversely affect neuronal development and did not cause congenital anomalies.


Subject(s)
Hippocampus/drug effects , Hypoglycemic Agents/toxicity , Maternal-Fetal Exchange , PPAR gamma/agonists , Thiazolidinediones/toxicity , Animals , Female , Hippocampus/pathology , Pregnancy , Rats , Rats, Wistar , Rosiglitazone
12.
Toxicol Lett ; 280: 151-158, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28822817

ABSTRACT

Rosiglitazone is an anti-diabetic agent that raised a major controversy over its cardiovascular adverse effects. There is in vivo evidence that Rosiglitazone promotes cardiac hypertrophy by PPAR-γ-independent mechanisms. However, whether Rosiglitazone directly alters hypertrophic growth in cardiac cells is unknown. Chromatin remodeling by histone post-translational modifications has emerged as critical for many cardiomyopathies. Based on these observations, this study was initiated to investigate the cardiac hypertrophic effect of Rosiglitazone in a cellular model of primary neonatal rat cardiomyocytes (NRCM). We assessed whether the drug alters cardiac hypertrophy and its relationship with histone H3 phosphorylation. Our study showed that Rosiglitazone is a mild pro-hypertrophic agent. Rosiglitazone caused a significant increase in the release of brain natriuretic peptide (BNP) into the cell media and also increased cardiomyocytes surface area and atrial natriuretic peptide (ANP) protein expression significantly. These changes correlated with increased cardiac phosphorylation of p38 MAPK and enhanced phosphorylation of H3 at serine 10 globally and at one cardiac hypertrophic gene locus. These results demonstrate that Rosiglitazone causes direct cardiac hypertrophy in NRCM and alters H3 phosphorylation status. They suggest a new mechanism of Rosiglitazone cardiotoxicity implicating chromatin remodeling secondary to H3 phosphorylation, which activate the fetal cardiac gene program.


Subject(s)
Cardiomegaly/chemically induced , Chromatin Assembly and Disassembly/drug effects , Fibrinolytic Agents/toxicity , Myocytes, Cardiac/drug effects , Thiazolidinediones/toxicity , Animals , Atrial Natriuretic Factor/metabolism , Epigenesis, Genetic , Female , Fibrinolytic Agents/administration & dosage , Gene Expression Regulation/drug effects , Histones/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Rosiglitazone , Thiazolidinediones/administration & dosage , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Toxicology ; 387: 1-9, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28645575

ABSTRACT

Drug-induced liver injury (DILI) is a leading cause of liver disease and a key safety factor during drug development. In addition to the initiation events of drug-specific hepatotoxicity, dysregulated immune responses have been proposed as major pathological events of DILI. Thus, there is a need for a reliable cell culture model with which to assess drug-induced immune reactions to predict hepatotoxicity for drug development. To this end, stem cell-derived hepatocytes have shown great potentials. Here we report that hepatocyte-like cells derived from human embryonic stem cells (hES-HLCs) can be used to evaluate drug-induced hepatotoxic immunological events. Treatment with acetaminophen significantly elevated the levels of inflammatory cytokines by hES-HLCs. Moreover, three human immune cell lines, Jurkat, THP-1, and NK92MI, were activated when cultured in conditioned medium obtained from acetaminophen-treated hES-HLCs. To further validate, we tested thiazolidinedione (TZD) class, antidiabetic drugs, including troglitazone withdrawn from the market because of severe idiosyncratic drug hepatotoxicity. We found that TZD drug treatment to hES-HLCs resulted in the production of pro-inflammatory cytokines and eventually associated immune cell activation. In summary, our study demonstrates for the first time the potential of hES-HLCs as an in vitro model system for assessment of drug-induced as well as immune-mediated hepatotoxicity.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Biological Assay , Cell Differentiation , Chemical and Drug Induced Liver Injury/etiology , Embryonic Stem Cells/drug effects , Hepatocytes/drug effects , Hypoglycemic Agents/toxicity , Thiazolidinediones/toxicity , Toxicity Tests/methods , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytokines/immunology , Cytokines/metabolism , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Jurkat Cells , Phenotype , Risk Assessment
14.
Toxicol Sci ; 157(2): 387-398, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28369597

ABSTRACT

Global gene expression profiling is useful for elucidating a drug's mechanism of action on the liver; however, such profiling in rats is not very sensitive for predicting human drug-induced liver injury, while dedifferentiated monolayers of primary human hepatocytes (PHHs) do not permit chronic drug treatment. In contrast, micropatterned cocultures (MPCCs) containing PHH colonies and 3T3-J2 fibroblasts maintain a stable liver phenotype for 4-6 weeks. Here, we used MPCCs to test the hypothesis that global gene expression patterns in stable PHHs can be used to distinguish clinical hepatotoxic drugs from their non-liver-toxic analogs and understand the mechanism of action prior to the onset of overt hepatotoxicity. We found that MPCCs treated with the clinical hepatotoxic/non-liver-toxic pair, troglitazone/rosiglitazone, at each drug's reported and non-toxic Cmax (maximum concentration in human plasma) for 1, 7, and 14 days displayed a total of 12, 269, and 628 differentially expressed genes, respectively, relative to the vehicle-treated control. Troglitazone modulated >75% of transcripts across pathways such as fatty acid and drug metabolism, oxidative stress, inflammatory response, and complement/coagulation cascades. Escalating rosiglitazone's dose to that of troglitazone's Cmax increased modulated transcripts relative to the lower dose; however, over half the identified transcripts were still exclusively modulated by troglitazone. Last, other hepatotoxins (nefazodone, ibufenac, and tolcapone) also induced a greater number of differentially expressed genes in MPCCs than their non-liver-toxic analogs (buspirone, ibuprofen, and entacapone) following 7 days of treatment. In conclusion, MPCCs allow evaluation of time- and dose-dependent gene expression patterns in PHHs treated chronically with analog drugs.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Fibroblasts/drug effects , Hepatocytes/drug effects , Liver/drug effects , Transcriptome/drug effects , 3T3 Cells , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/genetics , Chromans/toxicity , Coculture Techniques , Dose-Response Relationship, Drug , Fibroblasts/cytology , Gene Expression Profiling , Hepatocytes/cytology , Humans , Mice , Oxidative Stress/drug effects , Oxidative Stress/genetics , Primary Cell Culture , Rosiglitazone , Thiazolidinediones/toxicity , Toxicogenetics , Troglitazone
15.
Regul Toxicol Pharmacol ; 86: 25-32, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28202347

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors involved in the metabolism of lipids and carbohydrates. The exogenous ligands of these receptors are thiazolidinediones (TZDs), which are used to treat type 2 diabetes mellitus (DM2). However, drugs from this group produce adverse effects such as hepatic steatosis. Hence, the aim of this work was to design a set of small molecules that can activate the γ isoform of PPARs while minimizing the adverse effects. The derivatives were designed containing the polar head of TZD and an aromatic body, serving simultaneously as the body and tail. Two ligands were selected out of 130 tested. These compounds were synthesized in a solvent-free reaction and their physicochemical properties and toxicity were examined. Acute oral toxicity was determined by administering these compounds to female Wistar rats in increasing doses (as per the OECD protocol 425). The median lethal dose (LD50) of the compound substituted with a hydroxyl heteroatom was above 2000 mg/kg, and that of the compound substituted with halogens was 700-1400 mg/kg. The results suggest that the compounds can interact with PPARγ and elicit biological responses similar to other TZDs, but without showing adverse effects. The compounds will be subsequently evaluated in a DM2 animal model.


Subject(s)
Hypoglycemic Agents/toxicity , PPAR gamma/agonists , Thiazolidinediones/chemical synthesis , Thiazolidinediones/toxicity , Animals , Computer Simulation , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/chemical synthesis , Rats , Rats, Wistar
16.
Drug Des Devel Ther ; 10: 3947-3957, 2016.
Article in English | MEDLINE | ID: mdl-27980392

ABSTRACT

Hyperpigmentation caused by melanin overproduction is a major skin disorder in humans. Inhibition of tyrosinase, a key regulator of melanin production, has been used as an effective strategy to treat hyperpigmentation. In this study, we investigated the use of solid lipid nanoparticles (SLNs) as a highly effective and nontoxic means to deliver a newly synthesized potent tyrosinase inhibitor, MHY498, and to target melanocytes through the skin. MHY498-loaded SLNs (MHY-SLNs) were prepared by an oil-in-water emulsion solvent-evaporation method, and their morphological and physicochemical properties were characterized. MHY-SLNs showed a prolonged drug-release profile and higher skin permeation than that of MHY solution. In an in vivo evaluation of antimelanogenic activity, MHY-SLNs showed a prominent inhibitory effect against ultraviolet B-induced melanogenesis, resulting in no change in the skin color of C57BL/6 mouse, compared with that observed in an MHY solution-treated group and an untreated control group. The antimelanogenic effect of MHY-SLNs was further confirmed through Fontana-Masson staining. Importantly, MHY-SLNs did not induce any toxic effects in the L929 cell line. Overall, these data indicate that MHY-SLNs show promise in the topical treatment of hyperpigmentation.


Subject(s)
Drug Carriers , Enzyme Inhibitors/pharmacology , Lipids/chemistry , Melanins/metabolism , Nanoparticles , Skin Pigmentation/drug effects , Skin/drug effects , Thiazolidinediones/pharmacology , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Administration, Cutaneous , Animals , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Compounding , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Fibroblasts/drug effects , Fibroblasts/pathology , Kinetics , Mice, Inbred C57BL , Nanotechnology , Permeability , Skin/enzymology , Skin/radiation effects , Skin Absorption , Solubility , Technology, Pharmaceutical/methods , Thiazolidinediones/administration & dosage , Thiazolidinediones/chemistry , Thiazolidinediones/toxicity , Tyrosine 3-Monooxygenase/metabolism
17.
Eur J Med Chem ; 124: 608-621, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27614408

ABSTRACT

A series of new benzimidazole-thiazolidinedione hybrids has been synthesized and evaluated for their cytotoxic potential against a selected human cancer cell lines of prostate (PC-3 and DU-145), breast (MDA-MB-231), lung (A549) and a normal breast epithelial cells (MCF10A). Among the tested compounds, 11p exhibited promising cytotoxicity with IC50 value of 11.46 ± 1.46 µM on A549 lung cancer cell line and did not show significant toxicity on normal MCF10A cells. Lung cancer cells (A549) have been used to know the mechanism of cell growth inhibition and apoptosis inducing effect with compound 11p. The treatment of A549 cells with 11p showed typical apoptotic morphology like cell shrinkage, chromatin condensation and horseshoe shaped nuclei formation. Flow-cytometry analysis revealed the G2/M phase of cell cycle arrest in a dose dependent manner. Preliminary mechanistic studies suggested that the cell migration was inhibited through the disruption of F-actin protein. Acridine orange-ethidium bromide (AO-EB), DAPI, annexin V-FITC/propidium iodide, rhodamine-123 and MitoSOX assays suggested the induction of apoptosis in A549 cells by compound 11p.


Subject(s)
Apoptosis/drug effects , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Thiazolidinediones/chemical synthesis , Thiazolidinediones/pharmacology , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Benzimidazoles/chemistry , Benzimidazoles/toxicity , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Flow Cytometry , Humans , Inhibitory Concentration 50 , Molecular Structure , Neoplasms/classification , Neoplasms/drug therapy , Thiazolidinediones/chemistry , Thiazolidinediones/toxicity
18.
J Toxicol Sci ; 41(5): 605-15, 2016.
Article in English | MEDLINE | ID: mdl-27665770

ABSTRACT

Drug-induced liver injury (DILI) is one of the serious and frequent drug-related adverse events. This adverse event is a main reason for regulatory action pertaining to drugs, including restrictions in clinical indications and withdrawal from clinical trials or the marketplace. Idiosyncratic DILI especially has become a major clinical concern because of its unpredictable nature, frequent hospitalization, need for liver transplantation and high mortality. The estimation of the potential for compounds to induce idiosyncratic DILI is very difficult in non-clinical studies because the precise mechanism of idiosyncratic DILI is still unknown. Recently, many in vitro assays which indicate a possibility of the prediction of the idiosyncratic DILI have been reported. Among these, some in vitro assays focus on the effects of compounds on mitochondrial function and the apoptotic effects of compounds on human hepatocytes. In this study, we measured oxygen consumption rate (OCR) and caspase-3/7 activity as an endpoint of mitochondrial dysfunction and apoptosis, respectively, with human hepatocytes after treatment with compounds causing idiosyncratic DILI (troglitazone, leflunomide, ranitidine and diclofenac). Troglitazone and leflunomide decreased the OCR but did not affect caspase-3/7 activity. Ranitidine increased caspase-3/7 activity but did not affect the OCR. Diclofenac decreased the OCR and increased caspase-3/7 activity. Acetaminophen and ethanol, which are also hepatotoxicants but do not induce idiosyncratic DILI, did not affect the OCR or caspase-3/7 activity. These results indicate that a combination assay of mitochondrial dysfunction and apoptosis is useful for the estimation of potential risk of compounds to induce idiosyncratic DILI.


Subject(s)
Apoptosis/drug effects , Biological Assay , Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , Liver/drug effects , Mitochondria, Liver/drug effects , Toxicity Tests/methods , Acetaminophen/toxicity , Biomarkers/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chromans/toxicity , Diclofenac/toxicity , Dose-Response Relationship, Drug , Ethanol/toxicity , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Isoxazoles/toxicity , Leflunomide , Liver/metabolism , Liver/pathology , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Oxygen Consumption/drug effects , Primary Cell Culture , Ranitidine/toxicity , Risk Assessment , Thiazolidinediones/toxicity , Time Factors , Troglitazone
19.
Regul Toxicol Pharmacol ; 81: 155-161, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27569202

ABSTRACT

It has been reported that the major cause of mortality in diabetes is cardiovascular diseases and contribution of hypertension is significant in this context. Pioglitazone, a thiazolidinedione class of therapeutic agent is used to treat type 2 diabetes mellitus. Telmisartan, an angiotensin receptor blocker antihypertensive has been reported to have beneficial effect if co-administered with pioglitazone for the management of diabetes complications. The present research work aims to evaluate the safety/toxicity profile of this combination in rat model. The investigation was carried out after co-administering the drugs to the rats for 28 days at three dose levels of 50, 100 and 150 mg/kg covering low to high dose ranges. Various hematological and biochemical parameters were studied in addition to the histopathology of the major organs in order to evaluate the toxicity profile of the combination. Absence of mortality and histopathological changes as well as unaltered hematological and biochemical parameters was observed. This preliminary investigation concludes that the combination of pioglitazone and telmisartan can primarily be stated as safe in animals, even at the dose level which is several folds higher than the intended human dose. Thus, this combination can be explored in future to develop a rational therapy regimen to treat hypertensive diabetic patients.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/toxicity , Antihypertensive Agents/toxicity , Benzimidazoles/toxicity , Benzoates/toxicity , Hypoglycemic Agents/toxicity , Thiazolidinediones/toxicity , Toxicity Tests, Subchronic/methods , Administration, Oral , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Benzimidazoles/administration & dosage , Benzoates/administration & dosage , Dose-Response Relationship, Drug , Female , Hypoglycemic Agents/administration & dosage , Male , Pioglitazone , Polypharmacy , Rats, Wistar , Risk Assessment , Telmisartan , Thiazolidinediones/administration & dosage , Time Factors
20.
Drug Metab Lett ; 10(3): 206-212, 2016.
Article in English | MEDLINE | ID: mdl-27456668

ABSTRACT

BACKGROUND: LPSF/GQ-02 is a promising benzylidene thiazolidinedione that has demonstrated antidiabetic, antidyslipidemic, anti-atherosclerotic properties and can also treat non-alcoholic fatty liver disease. Despite all activity studies of the new compound, its pharmacokinetics are not yet described. OBJECTIVE: The aim of this study was to perform its first pharmacokinetic profile. METHODS: For this purpose a bioanalytical method for the quantitation of 5-(4- Chloro-benzylidene)-3-(4-methylbenzyl)-thiazolidine-2,4-dione (LPSF/GQ-02) was developed and validated. A Waters UPLC chromatographer using a BEH column (2.1x50mm, 1.7µm particle), mobile phase water:acetonitrile (20:80) was used. The range of calibration curve in plasma was 1.9 to 250 ng/mL with r = 0.9997. LPSF/GQ-02 stability was evaluated in rat plasma and buffers at pH 1.2 and 7.4. The pharmacokinetic assay was carried out in male Wistar rats weighing 250-300 g. The animals received LPSF/GQ-02 at 3 mg/kg by intravenous route. The animals were used to perform a preliminary safety study concerning the evaluation of liver and kidney biomarkers (ALT, AST, urea, creatinine). RESULTS: The obtained pharmacokinetic parameters were elimination half-life of 4.44 h, Cl of 8.00 L/h.kg, Vd of 45.60 L/kg and MRT of 3.79h. No difference was observed for the liver and kidney biomarkers. CONCLUSION: The intravenous pharmacokinetic parameters are in agreement with a good future posology, even though the plasma concentrations from oral administration were not quantifiable in a dose of 12 mg/kg. The preliminary safety study demonstrated no acute effect of the drug in liver and kidneys. The LPSF/GQ-02 is a new thiazolidinedione that should continue being evaluated for future clinical use.


Subject(s)
Chromatography, High Pressure Liquid/methods , Kidney/metabolism , Liver/metabolism , Thiazolidinediones/pharmacokinetics , Administration, Intravenous , Animals , Half-Life , Humans , Hydrogen-Ion Concentration , Male , Rats , Rats, Wistar , Thiazolidinediones/toxicity , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...