Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
Curr Pharm Biotechnol ; 21(9): 807-828, 2020.
Article in English | MEDLINE | ID: mdl-32321398

ABSTRACT

Drug-induced toxicity remains one of the leading causes of discontinuation of the drug candidate and post-marketing withdrawal. Thus, early identification of the drug candidates with the potential for toxicity is crucial in the drug development process. With the recent discovery of human- Induced Pluripotent Stem Cells (iPSC) and the establishment of the differentiation protocol of human iPSC into the cell types of interest, the differentiated cells from human iPSC have garnered much attention because of their potential applicability in toxicity evaluation as well as drug screening, disease modeling and cell therapy. In this review, we expanded on current information regarding the feasibility of human iPSC-derived cells for the evaluation of drug-induced toxicity with a focus on human iPSCderived hepatocyte (iPSC-Hep), cardiomyocyte (iPSC-CMs) and neurons (iPSC-Neurons). Further, we CSAHi, Consortium for Safety Assessment using Human iPS Cells, reported our gene expression profiling data with DNA microarray using commercially available human iPSC-derived cells (iPSC-Hep, iPSC-CMs, iPSC-Neurons), their relevant human tissues and primary cultured human cells to discuss the future direction of the three types of human iPSC-derived cells.


Subject(s)
Drug Discovery , Drug-Related Side Effects and Adverse Reactions/etiology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects , Neurons/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neurons/cytology , Neurons/metabolism
3.
Yakugaku Zasshi ; 138(6): 823-828, 2018.
Article in Japanese | MEDLINE | ID: mdl-29863054

ABSTRACT

 Use of the microelectrode array (MEA) system to record spontaneous neuron activity from networks of cultured neurons has potential as a good risk evaluation method for drug-induced seizure events. Spontaneous electrical activity in neural networks consists of action potential spikes and organized patterns of action potential bursts. In both potentiated rodent primary neurons and human induced pluripotent stem cell (iPSC)-derived neurons, an epileptogenic response pattern manifests as a synchronized burst from spatially separated neurons. Here, we delineate how to perform MEA experiments using cultured neurons, and how to analyze the MEA data to detect drug-induced seizurogenic abnormalities. Usually, a drug's effects, as shown by MEA data, include changes in spike frequency, inter-spike intervals (ISI), burst frequency, burst duration, spikes in a burst, etc. Subsequently, seizurogenic events are evidenced by changes in synchronized burst phenotypes from spatially separated multiple channels in an MEA probe, such as a change in the cross correlation of the spike events from all channels in an MEA probe, or a change in histogram from the sum of ISI for all channels in a probe, etc. We attempted to depict an epileptogenic marker using a histogram of the sum of spikes for all channels in an MEA probe. Verification of these metrics for drug induced abnormalities is ongoing in various collaboration organizations, including the Consortium for Safety Assessment using Human iPS Cells (CSAHi), iPS Non-clinical Experiments for the Nervous System (iNCENS). Collaboration networks for the utilization of iPSC-derived cells during drug development are also summarized here.


Subject(s)
Action Potentials/drug effects , Drug-Related Side Effects and Adverse Reactions , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Microelectrodes , Neurons/drug effects , Neurons/physiology , Seizures/chemically induced , Animals , Antineoplastic Agents/adverse effects , Cells, Cultured , Humans , Rats , Risk
4.
J Pharmacol Sci ; 136(4): 249-256, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29555184

ABSTRACT

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are expected to become a useful tool for proarrhythmia risk prediction in the non-clinical drug development phase. Several features including electrophysiological properties, ion channel expression profile and drug responses were investigated using commercially available hiPSC-CMs, such as iCell-CMs and Cor.4U-CMs. Although drug-induced arrhythmia has been extensively examined by microelectrode array (MEA) assays in iCell-CMs, it has not been fully understood an availability of Cor.4U-CMs for proarrhythmia risk. Here, we evaluated the predictivity of proarrhythmia risk using Cor.4U-CMs. MEA assay revealed linear regression between inter-spike interval and field potential duration (FPD). The hERG inhibitor E-4031 induced reverse-use dependent FPD prolongation. We next evaluated the proarrhythmia risk prediction by a two-dimensional map, which we have previously proposed. We determined the relative torsade de pointes risk score, based on the extent of FPD with Fridericia's correction (FPDcF) change and early afterdepolarization occurrence, and calculated the margins normalized to free effective therapeutic plasma concentrations. The drugs were classified into three risk groups using the two-dimensional map. This risk-categorization system showed high concordance with the torsadogenic information obtained by a public database CredibleMeds. Taken together, these results indicate that Cor.4U-CMs can be used for drug-induced proarrhythmia risk prediction.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Drug Discovery , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Biomarkers, Pharmacological , Cells, Cultured , Forecasting , Humans , Long QT Syndrome/chemically induced , Microelectrodes , Risk , Torsades de Pointes/chemically induced
5.
Toxicol Appl Pharmacol ; 329: 282-292, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28624441

ABSTRACT

Sunitinib malate (sunitinib) is an orally available, multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities. Although sunitinib is effective for the treatment of patients with gastrointestinal stromal tumor, advanced renal cell carcinoma, or pancreatic neuroendocrine tumor, adverse cardiac events associated with sunitinib administration have been reported. Here, we examined the effect of geldanamycin, an inhibitor of heat shock protein (Hsp) 90, on sunitinib-induced cytotoxicity in cardiomyocytes. First, we found that treatment with geldanamycin or other Hsp90 inhibitors (tanespimycin, ganetespib, or BIIB021) significantly attenuated sunitinib-induced cytotoxicity in rat H9c2 cardiomyocytes, suggesting a drug-class effect of Hsp90 inhibitors. We then examined the mechanisms underlying sunitinib-induced cytotoxicity and found that sunitinib induced autophagy in H9c2 cells and that pretreatment with geldanamycin inhibited the induction of autophagy by promoting degradation of the autophagy-related proteins Atg7, Beclin-1, and ULK1. Pharmacological assessment with autophagy inhibitors confirmed that geldanamycin attenuated the cytotoxicity of sunitinib by interfering with autophagy. In addition, we found that the molecular chaperone Hsp70, which is induced by geldanamycin, was not involved in the attenuation of sunitinib-induced cytotoxicity. Finally, to provide more clinically relevant data, we confirmed that geldanamycin attenuated sunitinib-induced cytotoxicity in human induced pluripotent stem cell-derived cardiomyocytes. Together, these data suggest that geldanamycin attenuates sunitinib-induced cytotoxicity in cardiomyocytes by inhibiting the autophagy pathway. Thus, the further investigation of combination or sequential treatment with an Hsp90 inhibitor and sunitinib is warranted as a potential strategy of attenuating the cardiotoxicity associated with sunitinib administration in the clinical setting.


Subject(s)
Antineoplastic Agents/toxicity , Autophagy/drug effects , Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Indoles/toxicity , Lactams, Macrocyclic/pharmacology , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/toxicity , Pyrroles/toxicity , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Cardiotoxicity , Cell Differentiation , Cell Line , Cell Lineage , Cytoprotection , Dose-Response Relationship, Drug , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA Interference , Rats , Signal Transduction/drug effects , Sunitinib , Transfection
6.
Article in English | MEDLINE | ID: mdl-28163191

ABSTRACT

INTRODUCTION: The use of multi-electrode arrays (MEA) in combination with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provides a promising method to predict comprehensive cardiotoxicity, including drug-induced QT prolongation and arrhythmia. We previously demonstrated that MEA in combination with hiPSC-CMs could provide a generalizable platform by using 7 reference drugs at 10 testing facilities. Using this approach, we evaluated responses to reference drugs that modulate a range of cardiac ion currents and have a range of arrhythmogenic effects. METHODS: We used the MEA system (MED64) and commercially available hiPSC-CMs (iCell cardiomyocytes) to evaluate drug effects on the beat rate, field potential duration (FPD), FPD corrected by Fridericia's formula (FPDc), and the incidence of arrhythmia-like waveforms. RESULTS: This assay detected the repolarization effects of Bay K8644, mibefradil, NS1643, levcromakalim, and ouabain; and the chronotropic effects of isoproterenol, ZD7288, and BaCl2. Chronotropy was also affected by K+ and Ca2+ current modulation. This system detected repolarization delays and the arrhythmogenic effects of quinidine, cisapride, thioridazine, astemizole, bepridil, and pimozide more sensitively than the established guinea pig papillary muscle action potential assay. It also predicted clinical QT prolongation by drugs with multiple ion channel effects (fluoxetine, amiodarone, tolterodine, vanoxerine, alfuzosin, and ranolazine). DISCUSSION: MEA in combination with hiPSC-CMs may provide a powerful method to detect various cardiac electrophysiological effects, QT prolongation, and arrhythmia during drug discovery. However, the data require careful interpretation to predict chronotropic effects and arrhythmogenic effects of candidate drugs with multiple ion channel effects.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiotoxins/pharmacology , Heart Rate/drug effects , Induced Pluripotent Stem Cells/drug effects , Ion Channels , Myocytes, Cardiac/drug effects , Arrhythmias, Cardiac/physiopathology , Cardiotonic Agents/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Heart Rate/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Ion Channels/agonists , Ion Channels/antagonists & inhibitors , Ion Channels/physiology , Myocytes, Cardiac/physiology
7.
Article in English | MEDLINE | ID: mdl-27646297

ABSTRACT

Recent increasing evidence suggests that the currently-used platforms in vitro IKr and APD, and/or in vivo QT assays are not fully predictive for TdP, and do not address potential arrhythmia (VT and/or VF) induced by diverse mechanisms of action. In addition, other cardiac safety liabilities such as functional dysfunction of excitation-contraction coupling (contractility) and structural damage (morphological damage to cardiomyocytes) are also major causes of drug attrition, but current in vitro assays do not cover all these liabilities. We organized the Consortium for Safety Assessment using Human iPS cells (CSAHi; http://csahi.org/en/), based on the Japan Pharmaceutical Manufacturers Association (JPMA), to verify the application of human iPS/ES cell-derived cardiomyocytes in drug safety evaluation. The main goal of the CSAHi HEART team has been to propose comprehensive screening strategies to predict a diverse range of cardiotoxicities by using recently introduced platforms (multi-electrode array (MEA), patch clamp, cellular impedance, motion field imaging [MFI], and Ca transient systems) while identifying the strengths and weaknesses of each. Our study shows that hiPS-CMs used in these platforms have pharmacological responses more relevant to humans in comparison with existent hERG, APD or Langendorff (MAPD/contraction) assays, and not only MEA but also other methods such as impedance, MFI, and Ca transient systems would offer paradigm changes of platforms for predicting drug-induced QT risk and/or arrhythmia or contractile dysfunctions. Furthermore, we propose a potential multi-parametric platform in which field potential (MEA)-Ca transient-contraction (MFI) could be evaluated simultaneously as an ideal novel platform for predicting a diversity of cardiac toxicities, namely whole effects on the excitation-contraction cascade.


Subject(s)
Action Potentials/drug effects , Arrhythmias, Cardiac/chemically induced , Drug-Related Side Effects and Adverse Reactions , Induced Pluripotent Stem Cells/drug effects , Models, Cardiovascular , Myocytes, Cardiac/drug effects , Cardiotoxicity , Cell Culture Techniques , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Induced Pluripotent Stem Cells/physiology , Microelectrodes , Myocytes, Cardiac/physiology , Pharmaceutical Preparations/administration & dosage
8.
J Pharmacol Toxicol Methods ; 84: 111-127, 2017.
Article in English | MEDLINE | ID: mdl-27956204

ABSTRACT

INTRODUCTION: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are anticipated to be a useful tool for conducting proarrhythmia risk assessments of drug candidates. However, a torsadogenic risk prediction paradigm using hiPSC-CMs has not yet been fully established. METHODS: Extracellular field potentials (FPs) were recorded from hiPSC-CMs using the multi-electrode array (MEA) system. The effects on FPs were evaluated with 60 drugs, including 57 with various clinical torsadogenic risks. Actual drug concentrations in medium were measured using the equilibrium dialysis method with a Rapid Equilibrium Dialysis device. Relative torsade de pointes (TdP) scores were determined for each drug according to the degree of FP duration prolongation and early afterdepolarization occurrence. The margins were calculated from the free concentration in medium and free effective therapeutic plasma concentration. Each drug's results were plotted on a two-dimensional map of relative TdP risk scores versus margins. RESULTS: Each drug was categorised as high, intermediate, or low risk based on its location within predefined areas of the two-dimensional map. We categorised 19 drugs as high risk; 18 as intermediate risk; and 17 as low risk. We examined the concordance between our categorisation of high and low risk drugs against the torsadogenic risk categorisation in CredibleMeds®. Our system demonstrated high concordance, as reflected in a sensitivity of 81%, specificity of 87%, and accuracy of 83%. DISCUSSION: These results indicate that our torsadogenic risk assessment is reliable and has a potential to replace the hERG assay for torsadogenic risk prediction, however, this system needs to be improved for the accurate of prediction of clinical TdP risk. Here, we propose a novel drug induced torsadogenic risk categorising system using hiPSC-CMs and the MEA system.


Subject(s)
Action Potentials/drug effects , Cardiotoxins/toxicity , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Torsades de Pointes/chemically induced , Action Potentials/physiology , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Risk Assessment , Torsades de Pointes/pathology , Torsades de Pointes/physiopathology
9.
PLoS One ; 11(12): e0167348, 2016.
Article in English | MEDLINE | ID: mdl-27923051

ABSTRACT

The aims of this study were to (1) characterize basic electrophysiological elements of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) that correspond to clinical properties such as QT-RR relationship, (2) determine the applicability of QT correction and analysis methods, and (3) determine if and how these in-vitro parameters could be used in risk assessment for adverse drug-induced effects such as Torsades de pointes (TdP). Field potential recordings were obtained from commercially available hiPSC-CMs using multi-electrode array (MEA) platform with and without ion channel antagonists in the recording solution. Under control conditions, MEA-measured interspike interval and field potential duration (FPD) ranged widely from 1049 to 1635 ms and from 334 to 527 ms, respectively and provided positive linear regression coefficients similar to native QT-RR plots obtained from human electrocardiogram (ECG) analyses in the ongoing cardiovascular-based Framingham Heart Study. Similar to minimizing the effect of heart rate on the QT interval, Fridericia's and Bazett's corrections reduced the influence of beat rate on hiPSC-CM FPD. In the presence of E-4031 and cisapride, inhibitors of the rapid delayed rectifier potassium current, hiPSC-CMs showed reverse use-dependent FPD prolongation. Categorical analysis, which is usually applied to clinical QT studies, was applicable to hiPSC-CMs for evaluating torsadogenic risks with FPD and/or corrected FPD. Together, this results of this study links hiPSC-CM electrophysiological endpoints to native ECG endpoints, demonstrates the appropriateness of clinical analytical practices as applied to hiPSC-CMs, and suggests that hiPSC-CMs are a reliable models for assessing the arrhythmogenic potential of drug candidates in human.


Subject(s)
Chromans/pharmacology , Cisapride/pharmacology , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/cytology , Piperidines/pharmacology , Pyridines/pharmacology , Sulfonamides/pharmacology , Cells, Cultured , Electrophysiological Phenomena/drug effects , Heart Rate/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Linear Models , Models, Cardiovascular , Torsades de Pointes/chemically induced , Torsades de Pointes/physiopathology
10.
J Biomol Screen ; 21(10): 1065-1074, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27572608

ABSTRACT

Neurotransmission mediated by acetylcholine receptors (AChRs) plays an important role in learning and memory functions in the hippocampus. Impairment of the cholinergic system contributes to Alzheimer's disease (AD), indicating the importance of AChRs as drug targets for AD. To improve the success rates for AD drug development, human cell models that mimic the target brain region are important. Therefore, we characterized the functional expression of nicotinic and muscarinic AChRs (nAChRs and mAChRs, respectively) in human hippocampal neurons differentiated from hippocampal neural stem/progenitor cells (HIP-009 cells). Intracellular calcium flux in 4-week differentiated HIP-009 cells demonstrated that the cells responded to acetylcholine, nicotine, and muscarine in a concentration-dependent manner (EC50 = 13.4 ± 0.5, 6.0 ± 0.4, and 35.0 ± 2.5 µM, respectively). In addition, assays using subtype-selective compounds revealed that major AD therapeutic target AChR subtypes-α7 and α4ß2 nAChRs, as well as M1 and M3 mAChRs-were expressed in the cells. Furthermore, neuronal network analysis demonstrated that potentiation of M3 mAChRs inhibits the spontaneous firing of HIP-009 neurons. These results indicate that HIP-009 cells are physiologically relevant for AD drug screening and hence are loadstars for the establishment of in vitro AD models.


Subject(s)
Alzheimer Disease/drug therapy , Cell Differentiation/genetics , Drug Delivery Systems/methods , Synaptic Transmission/drug effects , Acetylcholine/metabolism , Alzheimer Disease/genetics , Cell Differentiation/drug effects , Gene Expression Regulation, Developmental/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Humans , Muscarine/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Nicotine/metabolism , Patch-Clamp Techniques , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M3/antagonists & inhibitors , Receptor, Muscarinic M3/genetics , Receptors, Cholinergic/drug effects , Receptors, Cholinergic/genetics , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Stem Cells/cytology , Stem Cells/metabolism , Synaptic Transmission/genetics
11.
J Pharmacol Toxicol Methods ; 78: 93-102, 2016.
Article in English | MEDLINE | ID: mdl-26657830

ABSTRACT

INTRODUCTION: Drug-induced QT prolongation is a major safety issue during drug development because it may lead to lethal ventricular arrhythmias. In this study, we evaluated the utility of multi-electrode arrays (MEA) with human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) to predict drug-induced QT prolongation and arrhythmia. METHODS: Ten facilities evaluated the effects of 7 reference drugs (E-4031, moxifloxacin, flecainide, terfenadine, chromanol 293B, verapamil, and aspirin) using a MED64 MEA system with commercially available hiPS-CMs. Field potential duration (FPD), beat rate, FPD corrected by Fridericia's formula (FPDc), concentration inducing FPDc prolongation by 10% (FPDc10), and incidence of arrhythmia-like waveform were evaluated. RESULTS: The inter-facility variability of absolute values before drug application was similar to the intra-facility variability for FPD, beat rate, and FPDc. The inter-facility variability of FPDc10 for 5 reference drugs ranged from 1.8- to 5.8-fold. At all 10 facilities, E-4031, moxifloxacin, and flecainide prolonged FPDc and induced arrhythmia-like waveforms at concentrations 1.8- to 6.1-fold higher than their FPDc10. Terfenadine prolonged FPDc and induced beating arrest at 8.0 times the FPDc10. The average FPDc10 values for E-4031, moxifloxacin, and terfenadine were comparable to reported plasma concentrations that caused QT prolongation or Torsade de Pointes in humans. Chromanol 293B, a IKs blocker, also prolonged FPDc but did not induce arrhythmia-like waveforms, even at 7.4 times the FPDc10. In contrast, verapamil shortened FPDc and aspirin did not affect FPDc or FP waveforms. DISCUSSION: MEA with hiPS-CMs can be a generalizable method for accurately predicting both QT prolongation and arrhythmogenic liability in humans.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cell Culture Techniques/methods , Drug-Related Side Effects and Adverse Reactions , Induced Pluripotent Stem Cells/drug effects , Long QT Syndrome/chemically induced , Myocytes, Cardiac/drug effects , Arrhythmias, Cardiac/diagnosis , Congresses as Topic , Cryopreservation/methods , Drug-Related Side Effects and Adverse Reactions/diagnosis , Humans , Induced Pluripotent Stem Cells/physiology , Long QT Syndrome/diagnosis , Myocytes, Cardiac/physiology , Pharmaceutical Preparations/administration & dosage , Predictive Value of Tests
12.
Article in English | MEDLINE | ID: mdl-25910965

ABSTRACT

INTRODUCTION: Multi-electrode array (MEA) systems and human induced pluripotent stem (iPS) cell-derived cardiomyocytes are frequently used to characterize the electrophysiological effects of drug candidates for the prediction of QT prolongation and proarrhythmic potential. However, the optimal experimental conditions for obtaining reliable experimental data, such as high-pass filter (HPF) frequency and cell plating density, remain to be determined. METHODS: Extracellular field potentials (FPs) were recorded from iPS cell-derived cardiomyocyte sheets by using the MED64 and MEA2100 multi-electrode array systems. Effects of HPF frequency (0.1 or 1Hz) on FP duration (FPD) were assessed in the presence and absence of moxifloxacin, terfenadine, and aspirin. The influence of cell density on FP characteristics recorded through a 0.1-Hz HPF was examined. The relationship between FP and action potential (AP) was elucidated by simultaneous recording of FP and AP using a membrane potential dye. RESULTS: Many of the FP waveforms recorded through a 1-Hz HPF were markedly deformed and appeared differentiated compared with those recorded through a 0.1-Hz HPF. The concentration-response curves for FPD in the presence of terfenadine reached a steady state at concentrations of 0.1 and 0.3µM when a 0.1-Hz HPF was used. In contrast, FPD decreased at a concentration of 0.3µM with a characteristic bell-shaped concentration-response curve when a 1-Hz HPF was used. The amplitude of the first and second peaks in the FP waveform increased with increasing cell plating density. The second peak of the FP waveform roughly coincided with AP signal at 50% repolarization, and the negative deflection at the second peak of the FP waveform in the presence of E-4031 corresponded to early afterdepolarization and triggered activity. DISCUSSION: FP can be used to assess the QT prolongation and proarrhythmic potential of drug candidates; however, experimental conditions such as HPF frequency are important for obtaining reliable data.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Induced Pluripotent Stem Cells/cytology , Long QT Syndrome/chemically induced , Myocytes, Cardiac/drug effects , Action Potentials/drug effects , Arrhythmias, Cardiac/diagnosis , Aspirin/administration & dosage , Aspirin/toxicity , Dose-Response Relationship, Drug , Fluoroquinolones/administration & dosage , Fluoroquinolones/toxicity , Humans , Long QT Syndrome/diagnosis , Moxifloxacin , Piperidines/administration & dosage , Piperidines/toxicity , Pyridines/administration & dosage , Pyridines/toxicity , Terfenadine/administration & dosage , Terfenadine/toxicity
13.
J Pharmacol Sci ; 124(4): 494-501, 2014.
Article in English | MEDLINE | ID: mdl-24694996

ABSTRACT

A prospective comparison study across 3 independent research laboratories of a pure IKr blocker E-4031 was conducted by using the same batch of human iPS cell-derived cardiomyocytes in order to verify the utility and reliability of our original standard protocol. Field potential waveforms were recorded with a multi-electrode array system to measure the inter-spike interval and field potential duration. The effects of E-4031 at concentrations of 1 to 100 nM were sequentially examined every 10 min. In each facility, E-4031 significantly prolonged the field potential duration corrected by Fridericia's formula and caused early afterdepolarizations occasionally resulting in triggered activities, whereas it tended to decrease the rate of spontaneous contraction. These results were qualitatively and quantitatively consistent with previous non-clinical in vitro and in vivo studies as well as clinical reports. There were inter-facility differences in some absolute values of the results, which were not observed when the values were normalized as percentage change. Information described in this paper may serve as a guide when predicting the drug-induced repolarization delay and arrhythmias with this new technology of stem cells.


Subject(s)
Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Electrophysiologic Techniques, Cardiac/methods , Heart Rate/drug effects , Induced Pluripotent Stem Cells , Myocytes, Cardiac/cytology , Piperidines/pharmacology , Pyridines/pharmacology , Animals , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Depression, Chemical , Dose-Response Relationship, Drug , Humans , Prospective Studies
14.
Mol Cancer Ther ; 13(4): 823-32, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24448821

ABSTRACT

Many clinical cases of acquired resistance to the BRAF inhibitor vemurafenib have recently been reported. One of the causes of this acquired resistance is the BRAF downstream kinase point mutation MEK1-C121S. This mutation confers resistance to not only vemurafenib, but also to the allosteric MEK inhibitor selumetinib (AZD6244). Here, we investigated the pharmacologic activities and effectiveness of the novel MEK inhibitor E6201 against BRAF (v-raf murine sarcoma viral oncogene homolog B1)-V600E mutant melanoma harboring the MEK1-C121S mutation. A cell-free assay confirmed that E6201 is an ATP-competitive MEK inhibitor, meaning it has a different binding mode with MEK compared with allosteric MEK inhibitors. E6201 is more effective against BRAF-V600E mutant melanoma compared with BRAF wild-type melanoma based on MEK inhibition. We found that the acquired MEK1-C121S mutation in BRAF-V600E mutant melanoma conferred resistance to both vemurafenib and selumetinib but not E6201. The effectiveness of E6201 in this preclinical study is a result of its binding with MEK1 far from the C121S point mutation so the mutation is unable to influence the MAPK pathway inhibitory activity. These results support further clinical investigation of E6201.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Lactones/pharmacology , MAP Kinase Kinase 1/genetics , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Indoles/therapeutic use , Lactones/therapeutic use , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Melanoma/genetics , Mutagenesis, Site-Directed , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/therapeutic use , Vemurafenib
15.
J Pharmacol Toxicol Methods ; 69(2): 177-88, 2014.
Article in English | MEDLINE | ID: mdl-24296355

ABSTRACT

INTRODUCTION: Cardiac hypertrophy is a leading cause of many cardiovascular diseases, including heart failure, but its pathological mechanism is not fully understood. This study used highly purified human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes to produce an in vitro hypertrophy model and characterize its gene expression and electrophysiological properties. METHODS: For 7 days we cultured hiPSC-derived cardiomyocytes plated at high (2800-4800 cells/mm(2)) or low (500-1200 cells/mm(2)) cell density and assessed their cell size with confocal and fluorescence microscopy, their electrophysiological and pharmacological responses with multi-electrode array systems, and their gene expression patterns by using DNA microarray technology and quantitative PCR. We used quantitative PCR and Western blotting to compare the expression of potassium-channel genes between the hiPSC-derived cardiomyocytes and human fetal and adult hearts. RESULTS: The hiPSC-derived cardiomyocytes showed spontaneous beating and similar pattern of α-actinin molecules regardless of plating density. However, cells plated at low density had the following characteristics compared with those at high density: 1) significant enlargement in size; 2) significant increase or decrease in expression of the cardiac hypertrophy-characteristic genes NPPA, ATP2A2, ANKRD1 and MYL2 in accordance with the progression of hypertrophy; 3) significant reduction in responses to the inhibitors of cardiac slow delayed-rectifier K(+) current (IKs), chromanol 293B and HMR1556, in a cell-density-dependent manner; and 4) significant reduction in the expression of the KCNQ1 and KCNJ2 genes coding the K(+) ion channels conducting each IKs and cardiac inward rectifier outward K(+) current (IK1). DISCUSSION: The enlargement, hypertrophy-characteristic and potassium ion channels gene expression of hiPSC-derived cardiomyocytes suggest that low-density plating was sufficient to induce cardiac hypertrophy. This model may be useful in elucidating mechanisms underlying the onset and progress of cardiac hypertrophy, because these cells can be cultured for several weeks.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Culture Techniques , Electrophysiological Phenomena , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Adult , Cardiomegaly/genetics , Cell Separation , Electrodes , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Phenotype
16.
J Recept Signal Transduct Res ; 33(4): 224-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23697547

ABSTRACT

The effect of the intracerebroventricular (i.c.v.) injection of relaxin-3 (RLX3) was evaluated using anxiety-related behavioral tests in rats. RLX3-injected animals showed normal locomotion activity in a habituated environment and declined anxiety cognition in the elevated plus maze test and the shock probe-burying test. The measurement of spontaneous locomotor activity in a novel environment also suggested that RLX3 reduced the stress response. To elucidate the regulatory mechanisms of the downstream signaling pathways underlying RLX3 activity and its relation to anxiolytic and hyperphagic behavior phenotypes, RLX3-i.c.v.-injected rat hypothalamic responses were examined using a microarray analysis. Ingenuity Pathway Analysis software listed the phenotype-relating genes and they showed characteristic expression patterns in the rat hypothalamus. When peptidome data sets for the same listed genes was analyzed using a semi-quantitative approach, the expressions of two neuropeptides were found to have increased. One of these neuropeptides, oxytocin (Oxt), exhibited increased expression in both the microarray and the peptidomic analysis, and a Western blot analysis validated the mass spectrometry results. A cross-omics data analysis is useful for predicting downstream signaling pathways, and the anxiolytic-like behavior of RLX3 may be mediated by an oxytocin signaling pathway in rats. These results suggest that RLX3 acts as an anxiolytic peptide and that the downstream pathways mediated by its receptors may be potential candidates for the treatment of anxieties in the future.


Subject(s)
Anxiety/drug therapy , Behavior, Animal/drug effects , Nerve Tissue Proteins/metabolism , Relaxin/metabolism , Stress, Physiological/drug effects , Animals , Anxiety/physiopathology , Behavior, Animal/physiology , Hypothalamus/metabolism , Injections, Intraventricular , Maze Learning , Microarray Analysis , Nerve Tissue Proteins/administration & dosage , Neuropeptides/isolation & purification , Neuropeptides/metabolism , Oxytocin/metabolism , Rats , Relaxin/administration & dosage , Signal Transduction
17.
Hum Exp Toxicol ; 32(10): 1028-37, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23424208

ABSTRACT

Channels responsible for slowly activating delayed-rectifier potassium current (I(Ks)) are composed of KCNQ1 and KCNE1 subunits, and these channels play a role in the repolarization of cardiac action potentials. Recently, we showed that the antihyperlipidemic drug probucol, which induces QT prolongation, decreases the I(Ks) after 24-h treatment. In the present study, we investigated the effects of three cholesterol-lowering agents (probucol, an enhancer of cholesterol efflux; simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor; and triparanol, a 3ß-hydroxysterol-▵24-reductase inhibitor) on cholesterol synthesis, the KCNQ1 current (I KCNQ1), and the I(Ks) to clarify the differences in the modes of action of these agents on the I(Ks). Probucol did not inhibit cholesterol synthesis and had no effect on I KCNQ1, while I(Ks) decreased after 24-h treatment. Simvastatin inhibited cholesterol synthesis and decreased I KCNQ1 and I(Ks). Additionally, the activation kinetics of I(Ks) became faster, compared with that of control I(Ks). Triparanol inhibited cholesterol synthesis but did not reduce I KCNQ1 and I(Ks). However, the activation kinetics of I(Ks) became faster. Our data indicated that the mechanism by which probucol inhibits I(Ks) was not mediated by the inhibition of cholesterol synthesis but depended on an interaction with the KCNQ1/KCNE1 complex. Meanwhile, the reduction in cholesterol induced by simvastatin and triparanol is one of the mechanisms that affects the kinetics of I(ks).


Subject(s)
Anticholesteremic Agents/pharmacology , Potassium Channels, Voltage-Gated/physiology , Probucol/pharmacology , Simvastatin/pharmacology , Triparanol/pharmacology , Animals , CHO Cells , Cholesterol/metabolism , Cricetulus
18.
J Cardiovasc Pharmacol ; 59(4): 377-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22189896

ABSTRACT

Indirect effects of drugs on ion channel expression levels on plasma membrane are focused as the cause of QT prolongation, and we explored the chronic effects of QT-prolonging drugs on the slow component of the delayed-rectifier potassium current (IKs). Chinese Hamster Ovary cells expressing IKs channels were constructed by transfecting KCNQ1/KCNE1 genes, and the IKs values were measured using IonWorks Quattro in the population patch-clamp mode. After 24 hours of treatment with IKs blockers (HMR1556, L-768673, or chromanol 293B) or human Ether-à-go-go related gene channel trafficking inhibitors (amiodarone,17-AAG, brefeldin A, pentamidine, thioridazine, or probucol), brefeldin A, pentamidine, and probucol decreased IKs. Probucol, which is a cholesterol-lowering drug and clinically reported to cause QT prolongation, potently inhibited the IKs in a concentration-dependent manner, with a half maximal inhibitory concentration of 149.1 nM. A reduction in the IKs by 1 µM of probucol was observed beginning 2 hours after treatment, and the current was reduced by about 80% at 24 hours. The activation and deactivation time constants of residual IKs currents became faster compared with that in the vehicle-treatment group. Acute application of probucol did not directly inhibit IKs channels at concentrations of up to 10 µM. Western blotting analysis indicated the reduction of multimeric complex of KCNQ1 proteins by probucol treatment but not monomeric form. These results suggest that chronic probucol treatment may contribute to QT prolongation in humans by decreasing the functional IKs channel complexes.


Subject(s)
Anticholesteremic Agents/toxicity , Long QT Syndrome/chemically induced , Potassium Channels, Voltage-Gated/drug effects , Probucol/toxicity , Animals , Anticholesteremic Agents/administration & dosage , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/genetics , Humans , KCNQ1 Potassium Channel/genetics , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Probucol/administration & dosage , Time Factors , Transfection
19.
J Biol Chem ; 285(43): 33294-33306, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20720005

ABSTRACT

One of the family of voltage-gated calcium channels (VGCC), the N-type Ca(2+) channel, is located predominantly in neurons and is associated with a variety of neuronal responses, including neurodegeneration. A precise mechanism for how the N-type Ca(2+) channel plays a role in neurodegenerative disease, however, is unknown. In this study, we immunized N-type Ca(2+) channel α(1B)-deficient (α(1B)(-/-)) mice and their wild type (WT) littermates with myelin oligodendrocyte glycoprotein 35-55 and analyzed the progression of experimental autoimmune encephalomyelitis (EAE). The neurological symptoms of EAE in the α(1B)(-/-) mice were less severe than in the WT mice. In conjunction with these results, sections of the spinal cord (SC) from α(1B)(-/-) mice revealed a reduction in both leukocytic infiltration and demyelination compared with WT mice. No differences were observed in the delayed-type hypersensitivity response, spleen cell proliferation, or cytokine production from splenocytes between the two genotypes. On the other hand, Western blot array analysis and RT-PCR revealed that a typical increase in the expression of MCP-1 in the SC showed a good correlation with the infiltration of leukocytes into the SC. Likewise, immunohistochemical analysis showed that the predominant source of MCP-1 was activated microglia. The cytokine-induced production of MCP-1 in primary cultured microglia from WT mice was significantly higher than that from α(1B)(-/-) mice and was significantly inhibited by a selective N-type Ca(2+) channel antagonist, ω-conotoxin GVIA or a withdrawal of extracellular Ca(2+). These results suggest that the N-type Ca(2+) channel is involved in the pathogenesis of EAE at least in part by regulating MCP-1 production by microglia.


Subject(s)
Calcium Channels, N-Type/metabolism , Chemokine CCL2/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glycoproteins/metabolism , Microglia/metabolism , Peptide Fragments/metabolism , Spinal Cord/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/genetics , Chemokine CCL2/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Glycoproteins/genetics , Leukocytes/metabolism , Leukocytes/pathology , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Microglia/pathology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/genetics , Spinal Cord/pathology , omega-Conotoxin GVIA/pharmacology
20.
Curr Pharm Biotechnol ; 10(2): 261-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19199960

ABSTRACT

Using gene knockout mice of particular genes is one of the most effective methods in conducting successful study on the mode of action of target gene products in targeted organs. So called the knockout technology is now a powerful tool that can lead us to find clear understanding on difficult questions such as the effects of full antagonist against target molecules. Cacna1b (alpha(1B)) gene knockout mouse was generated to study mechanisms of N-type calcium (Ca(2+)) channel. The model was able to overcome physiological obstacles in studies of N-type Ca(2+) channel selective blockers, such as unspecific binding to structurally similar molecules, and failed distribution to targeted organs. In the case of N-type Ca(2+) channel studies, knockout technology was successfully applied to various cardiovascular, sympathetic, nociceptive, sleep-awake cycles, metabolic and neurodegenerative experiments using homozygous mutants of the alpha(1B) gene that turned out to be viable. These studies were able to confirm not only the predicted phenotypes, but were able to present completely unexpected phenotypes that are great interest for future study. Thus the outputs from the knockout mouse studies lead to gain the proof of concept as a drug for specific inhibitors of the gene products and enabled us to make further prediction of side-effects of these inhibitors in the drug discovery and development process.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/physiology , Drug Design , Gene Targeting , Models, Biological , Animals , Calcium Channel Blockers/adverse effects , Calcium Channels, N-Type/metabolism , Drug Evaluation, Preclinical/methods , Mice , Mice, Knockout , Predictive Value of Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...