Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Sci Rep ; 14(1): 14109, 2024 06 19.
Article En | MEDLINE | ID: mdl-38898080

Developing a reliable method to predict thrombocytopenia is imperative in drug discovery. Here, we establish an assay using a microphysiological system (MPS) to recapitulate the in-vivo mechanisms of platelet aggregation and adhesion. This assay highlights the role of shear stress on platelet aggregation and their interactions with vascular endothelial cells. Platelet aggregation induced by soluble collagen was detected under agitated, but not static, conditions using a plate shaker and gravity-driven flow using MPS. Notably, aggregates adhered on vascular endothelial cells under gravity-driven flow in the MPS, and this incident increased in a concentration-dependent manner. Upon comparing the soluble collagen-induced aggregation activity in platelet-rich plasma (PRP) and whole blood, remarkable platelet aggregate formation was observed at concentrations of 30 µg/mL and 3 µg/mL in PRP and whole blood, respectively. Moreover, ODN2395, an oligonucleotide, induced platelet aggregation and adhesion to vascular endothelial cells. SYK inhibition, which mediated thrombogenic activity via glycoprotein VI on platelets, ameliorated platelet aggregation in the system, demonstrating that the mechanism of platelet aggregation was induced by soluble collagen and oligonucleotide. Our evaluation system partially recapitulated the aggregation mechanisms in blood vessels and can contribute to the discovery of safe drugs to mitigate the risk of thrombocytopenia.


Blood Platelets , Platelet Aggregation , Thrombocytopenia , Platelet Aggregation/drug effects , Humans , Thrombocytopenia/chemically induced , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen/metabolism , Collagen/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Platelet Adhesiveness/drug effects , Syk Kinase/metabolism , Syk Kinase/antagonists & inhibitors , Platelet-Rich Plasma/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Microphysiological Systems
2.
Toxicol Lett ; 396: 11-18, 2024 May 15.
Article En | MEDLINE | ID: mdl-38631510

Mitochondrial fatty acid oxidation (mtFAO) plays an important role in hepatic energy metabolism. Severe mtFAO injury leads to nonalcoholic fatty liver disease (NAFLD) and liver failure. Several drugs have been withdrawn owing to safety issues, such as induction of fatty liver disease through mtFAO disruption. For instance, the antimicrobial triclocarban (TCC), an environmental contaminant that was removed from the market due to its unknown safety in humans, induces NAFLD in rats and promotes hepatic FAO in mice. Therefore, there are no consistent conclusions regarding the effects of TCC on FAO and lipid droplet accumulation. We hypothesized that TCC induces lipid droplet accumulation by inhibiting mtFAO in human hepatocytes. Here, we evaluated mitochondrial respiration in HepaRG cells to investigate the effects of TCC on fatty acid-driven oxidation in cells, electron transport chain parameters, lipid droplet accumulation, and antioxidant genes. The results suggest that TCC increases oxidative stress gene expression (GCLM, p62, HO-1, and NRF2) through lipid droplet accumulation via mtFAO inhibition in HepaRG cells. The results of the present study provide further insights into the effect of TCC on human NAFLD through mtFAO inhibition, and further in vivo studies could be used to validate the mechanisms.


Carbanilides , Fatty Acids , Hepatocytes , Lipid Droplets , Oxidation-Reduction , Oxidative Stress , Humans , Oxidative Stress/drug effects , Carbanilides/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipid Droplets/metabolism , Lipid Droplets/drug effects , Fatty Acids/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Cell Line , Mitochondria/drug effects , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Lipid Metabolism/drug effects
3.
Toxicol Res (Camb) ; 12(5): 751-755, 2023 Oct.
Article En | MEDLINE | ID: mdl-37915470

Reliable fluid biomarkers for evaluating neurotoxicity have yet to be established. However, recent studies have reported neurofilament light chain as a fluid biomarker of several neurodegenerative disorders. In this study, we investigated changes in the cerebrospinal fluid and plasma levels of neurofilament light chain in mice treated with trimethyltin as a neurotoxicant. Trimethyltin diluted with saline was administered by intraperitoneal injection to mice at dose levels of 0 (vehicle control), 1.0, and 2.6 mg/kg body weight (dosage volume: 10 mL/kg). At 3 or 7 days after administration, animals were euthanized by exsanguination under 2-3% isoflurane inhalation anesthesia. Increased neurofilament light chain levels in both the cerebrospinal fluid and plasma were observed in animals from the trimethyltin 2.6 mg/kg body weight group, which indicated the brain lesions including neuronal cell death. Animals from the trimethyltin 1.0 mg/kg body weight group exhibited changes neither in neurofilament light chain levels in the cerebrospinal fluid and plasma nor in the histopathology of the brain at any time point. These data indicate that plasma neurofilament light chain can serve as a useful peripheral biomarker for detecting brain lesions such as neuronal necrosis in mice.

4.
J Toxicol Pathol ; 36(4): 199-204, 2023 Oct.
Article En | MEDLINE | ID: mdl-37868118

Determining the optimal timing for histopathological examination following exposure to a test article is crucial for assessing neurotoxicity. However, no study has focused on identifying an ideal dataset to define the optimal timing for histopathological examination of central nervous system (CNS) toxicity in monkeys. Therefore, this study aimed to define a predictive endpoint that would guide us in selecting the optimal timing for histopathological examination of CNS toxicity in monkeys. Four cynomolgus monkeys were administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intravenously at a dosage of 0.6 mg/kg twice at 1-week intervals. Necropsies were performed 1 week after the final dose. The Parkinsonian rating (PR) score and temporal changes in neurofilament light chain and glial fibrillary acidic protein concentrations in the cerebrospinal fluid (CSF) and serum were evaluated and compared with the histopathological findings in the brain. The PR score of all animals administered MPTP increased from days 10 to 11, with some degree of individual variability. Microscopically, all animals showed axonal swelling and vacuolation, with or without microgliosis in the nigrostriatal bundle. However, substantial neurodegenerative findings were observed only in animals with high PR scores at necropsy. A slight increase in CSF biomarker levels at necropsy was also observed in animals with high PR scores. However, their correlation with microscopic findings in these animals was unclear. These data suggest that comprehensive clinical observations, such as PR score alone or combined with other CSF biomarkers, could be further evaluated as potential indicators for triggering anatomic CNS evaluations in monkeys following toxic insults.

5.
J Toxicol Pathol ; 36(2): 145-149, 2023 Apr.
Article En | MEDLINE | ID: mdl-37101960

Neurofilament light chain (NfL) has recently been used as a biomarker of neurodegeneration. Although cerebrospinal fluid (CSF) NfL levels are hypothesized to affect blood NfL levels, whether blood NfL levels change independently of the CSF during peripheral nerve injury remains unclear. Thus, we evaluated the nervous tissues histopathology and serum and CSF NfL levels in partial sciatic nerve-ligated rats at 6 h and one, three, or seven days after the surgery. Sciatic and tibial nerve fiber damage was observed at 6 h after the surgery, and peaked at three days postoperatively. The serum NfL levels peaked 6 h to one day after ligation, but they tended to return to the normal seven days after ligation. However, the CSF NfL levels were unchanged throughout the study period. In conclusion, the comparative evaluation of serum and CSF NfL levels can provide useful information as biomarkers of nerve tissue damage and its distribution.

6.
Toxicol In Vitro ; 89: 105569, 2023 Jun.
Article En | MEDLINE | ID: mdl-36801361

In this study, we investigated thrombocytopenia caused by cholesterol-conjugated antisense oligonucleotides (Chol-ASO). First, we evaluated platelet activation induced by Chol-ASO in mice by flow cytometry after administration of platelet-rich plasma (PRP). An increase in the number of large particle-size events with platelet activation was detected in the Chol-ASO-treated group. In a smear study, numerous platelets were observed to attach to nucleic acid-containing aggregates. A competition binding assay showed that the conjugation of cholesterol to ASOs increased their affinity for glycoprotein VI. Platelet-free plasma was then mixed with Chol-ASO to form aggregates. The assembly of Chol-ASO was confirmed by dynamic light scattering measurements in the concentration range in which the formation of aggregates with plasma components was observed. In conclusion, the mechanism by which Chol-ASOs causes thrombocytopenia is proposed to be as follows: (1) Chol-ASOs form polymers, (2) the nucleic acid portion of the polymers interacts with plasma proteins and platelets, which cross-links them to form aggregates, and (3) platelets bound to aggregates become activated, resulting in platelet aggregation, leading to a decrease in platelet count in vivo. The details of the mechanism revealed in this study could contribute to creating safer oligonucleotide therapies without the risk of thrombocytopenia.


Oligonucleotides, Antisense , Thrombocytopenia , Animals , Mice , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/pharmacology , Cholesterol , Thrombocytopenia/chemically induced , Blood Platelets/metabolism
7.
Front Genet ; 12: 767621, 2021.
Article En | MEDLINE | ID: mdl-34790228

Organoids are three-dimensional structures fabricated in vitro from pluripotent stem cells or adult tissue stem cells via a process of self-organization that results in the formation of organ-specific cell types. Human organoids are expected to mimic complex microenvironments and many of the in vivo physiological functions of relevant tissues, thus filling the translational gap between animals and humans and increasing our understanding of the mechanisms underlying disease and developmental processes. In the last decade, organoid research has attracted increasing attention in areas such as disease modeling, drug development, regenerative medicine, toxicology research, and personalized medicine. In particular, in the field of toxicology, where there are various traditional models, human organoids are expected to blaze a new path in future research by overcoming the current limitations, such as those related to differences in drug responses among species. Here, we discuss the potential usefulness, limitations, and future prospects of human liver, heart, kidney, gut, and brain organoids from the viewpoints of predictive toxicology research and drug development, providing cutting edge information on their fabrication methods and functional characteristics.

8.
Toxicol Sci ; 185(1): 10-18, 2021 12 28.
Article En | MEDLINE | ID: mdl-34677616

Neurotoxicity is a principal concern in nonclinical drug development. However, standardized and universally accepted fluid biomarkers for evaluating neurotoxicity are lacking. Increasing clinical evidence supports the potential use of neurofilament light (NfL) chain as a biomarker of several neurodegenerative diseases; therefore, we investigated changes in the cerebrospinal fluid (CSF) and serum levels of NfL in Sprague Dawley rats treated with central nervous system (CNS) toxicants (trimethyltin [TMT, 10 mg/kg po, single dose], kainic acid [KA, 12 mg/kg sc, single dose], MK-801 [1 mg/kg sc, single dose]), and a peripheral nervous system (PNS) toxicant (pyridoxine, 1200 mg/kg/day for 3 days). Animals were euthanized 1 (day 2), 3 (day 4), or 7 days after administration (day 8). Increased serum NfL was observed in TMT- and KA-treated animals, which indicated neuronal cell death in the brain on days 2, 4, and/or 8. MK-801-treated animals exhibited no changes in the serum and CSF levels of NfL and no histopathological changes in the brain at any time point. Pyridoxine-induced chromatolysis of the dorsal root ganglion on day 2 and degeneration of peripheral nerve fiber on day 4; additionally, serum NfL was increased. A strong correlation was observed between the serum and CSF levels of NfL and brain lesions caused by TMT and KA, indicating that NfL could be a useful biomarker for detecting CNS toxicity. Additionally, PNS changes were correlated with serum NfL levels. Therefore, serum NfL could serve as a useful peripheral biomarker for detecting both CNS and PNS toxicity in rats.


Intermediate Filaments , Peripheral Nerves , Animals , Biomarkers , Central Nervous System/metabolism , Intermediate Filaments/metabolism , Peripheral Nerves/metabolism , Rats , Rats, Sprague-Dawley
9.
BMC Pharmacol Toxicol ; 22(1): 55, 2021 10 06.
Article En | MEDLINE | ID: mdl-34610839

BACKGROUND: Sunitinib is known to cause cardiotoxicity in clinical settings. However, among sunitinib-treated patients experiencing adverse cardiac events, decreased cardiac function was reportedly reversible in > 50% of the patients. We previously showed that anti-cancer drugs such as sunitinib cause marked sarcomere disruption in human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), and the extent of sarcomere disruption can be used to predict drug-induced cardiotoxicity in humans. The aim of this study is to investigate whether the reversibility of sunitinib-induced cardiac events in clinical settings can be mimicked in vitro, and to examine the molecular mechanism responsible for sunitinib-induced cardiotoxicity focusing on the Hippo pathway. METHODS: iPSC-CMs were stimulated with sunitinib for 72 h and the morphology of sarcomere structures were analyzed by high-content analysis before and after sunitinib washout. To examine the involvement of the Hippo pathway in the sunitinib-induced sarcomere disruption, the extent of nuclear localization of YAP1 (yes-associated protein 1, a Hippo signaling target) was determined. iPSC-CMs were also stimulated with sunitinib and a small molecule inhibitor of the Hippo pathway, XMU-MP-1 and sarcomere structures were analyzed. RESULTS: We observed a spontaneous recovery in cardiac sarcomeres in iPSC-CMs that were significantly disrupted by sunitinib treatment after a 72 h or 144 h washout of sunitinib. The extent of nuclear localization of YAP1 was significantly reduced after sunitinib stimulation and tended to return to normal levels after drug washout. Simultaneous stimulation of iPSC-CM with sunitinib and XMU-MP-1 suppressed the sunitinib-induced disruption of sarcomeres. CONCLUSIONS: These results indicate that iPSC-CMs have the ability to recover from sunitinib-induced sarcomere disruption, and the Hippo pathway plays a role in the process of sunitinib-induced disruption of sarcomere and its recovery. Inhibition of the Hippo pathway may help to develop a co-medication strategy for mitigating the risk of sunitinib-induced adverse cardiac events.


Antineoplastic Agents/administration & dosage , Hippo Signaling Pathway/drug effects , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/administration & dosage , Sulfonamides/administration & dosage , Sunitinib/administration & dosage , Cells, Cultured , Humans , Myocytes, Cardiac/metabolism , Sarcomeres/drug effects , YAP-Signaling Proteins/metabolism
10.
Toxicol In Vitro ; 74: 105159, 2021 Aug.
Article En | MEDLINE | ID: mdl-33823239

Here, we established a high-throughput in vitro assay system to predict reactive metabolite (RM) formation. First, we performed the glutathione (GSH) consumption assay to monitor GSH levels as an index of RM formation potential using HepaRG cells pretreated with 500 µM D,L-buthionine-(S,R)-sulfoximine (BSO) and then treated with ticlopidine and diclofenac. Both drugs, under GSH-reduced conditions, significantly decreased relative cellular GSH content by 70% and 34%, respectively, compared with that in cells not pretreated with BSO. Next, we examined the correlation between GSH consumption and covalent binding assays; the results showed good correlation (correlation coefficient = 0.818). We then optimized the test compound concentration for evaluating RM formation potential using 76 validation compound sets, and the highest sensitivity (53%) was observed at 100 µM. Finally, using HepG2 cells, PXB-cells, and human primary hepatocytes, we examined the cell types suitable for evaluating RM formation potential. The expression of CYP3A4 was highest in HepaRG cells, suggesting the highest sensitivity (56.4%) of the GSH consumption assay. Moreover, a co-culture model of PXB-cells and HepaRG cells showed high sensitivity (72.7%) with sufficient specificity (85.7%). Thus, the GSH consumption assay can be used to effectively evaluate RM formation potential in the early stages of drug discovery.


Activation, Metabolic , Glutathione/metabolism , High-Throughput Screening Assays , Aspirin/toxicity , Buthionine Sulfoximine/pharmacology , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Cytochrome P-450 Enzyme System/metabolism , Diclofenac/toxicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Microsomes, Liver/metabolism , Ticlopidine/toxicity
11.
Gastroenterology ; 160(3): 831-846.e10, 2021 02.
Article En | MEDLINE | ID: mdl-33039464

BACKGROUND & AIMS: Preclinical identification of compounds at risk of causing drug induced liver injury (DILI) remains a significant challenge in drug development, highlighting a need for a predictive human system to study complicated DILI mechanism and susceptibility to individual drug. Here, we established a human liver organoid (HLO)-based screening model for analyzing DILI pathology at organoid resolution. METHODS: We first developed a reproducible method to generate HLO from storable foregut progenitors from pluripotent stem cell (PSC) lines with reproducible bile transport function. The qRT-PCR and single cell RNA-seq determined hepatocyte transcriptomic state in cells of HLO relative to primary hepatocytes. Histological and ultrastructural analyses were performed to evaluate micro-anatomical architecture. HLO based drug-induced liver injury assays were transformed into a 384 well based high-speed live imaging platform. RESULTS: HLO, generated from 10 different pluripotent stem cell lines, contain polarized immature hepatocytes with bile canaliculi-like architecture, establishing the unidirectional bile acid transport pathway. Single cell RNA-seq profiling identified diverse and zonal hepatocytic populations that in part emulate primary adult hepatocytes. The accumulation of fluorescent bile acid into organoid was impaired by CRISPR-Cas9-based gene editing and transporter inhibitor treatment with BSEP. Furthermore, we successfully developed an organoid based assay with multiplexed readouts measuring viability, cholestatic and/or mitochondrial toxicity with high predictive values for 238 marketed drugs at 4 different concentrations (Sensitivity: 88.7%, Specificity: 88.9%). LoT positively predicts genomic predisposition (CYP2C9∗2) for Bosentan-induced cholestasis. CONCLUSIONS: Liver organoid-based Toxicity screen (LoT) is a potential assay system for liver toxicology studies, facilitating compound optimization, mechanistic study, and precision medicine as well as drug screening applications.


Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , High-Throughput Screening Assays/methods , Liver/drug effects , Organoids/drug effects , Cell Line , Chemical and Drug Induced Liver Injury/pathology , Drug Evaluation, Preclinical/methods , Hepatocytes/pathology , Humans , Liver/cytology , Liver/pathology , Organoids/pathology , Pluripotent Stem Cells/cytology , Toxicity Tests, Acute/methods
12.
Nat Med ; 26(10): 1541-1548, 2020 10.
Article En | MEDLINE | ID: mdl-32895570

Drug-induced liver injury (DILI) is a leading cause of termination in drug development programs and removal of drugs from the market; this is partially due to the inability to identify patients who are at risk1. In this study, we developed a polygenic risk score (PRS) for DILI by aggregating effects of numerous genome-wide loci identified from previous large-scale genome-wide association studies2. The PRS predicted the susceptibility to DILI in patients treated with fasiglifam, amoxicillin-clavulanate or flucloxacillin and in primary hepatocytes and stem cell-derived organoids from multiple donors treated with over ten different drugs. Pathway analysis highlighted processes previously implicated in DILI, including unfolded protein responses and oxidative stress. In silico screening identified compounds that elicit transcriptomic signatures present in hepatocytes from individuals with elevated PRS, supporting mechanistic links and suggesting a novel screen for safety of new drug candidates. This genetic-, cellular-, organoid- and human-scale evidence underscored the polygenic architecture underlying DILI vulnerability at the level of hepatocytes, thus facilitating future mechanistic studies. Moreover, the proposed 'polygenicity-in-a-dish' strategy might potentially inform designs of safer, more efficient and robust clinical trials.


Chemical and Drug Induced Liver Injury/genetics , Multifactorial Inheritance , Polymorphism, Single Nucleotide , Alleles , Benzofurans/therapeutic use , Case-Control Studies , Cells, Cultured , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/epidemiology , Cohort Studies , Datasets as Topic/statistics & numerical data , Gene Expression Profiling , Gene Frequency , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Microarray Analysis , Multifactorial Inheritance/genetics , Sulfones/therapeutic use
13.
J Pharmacol Toxicol Methods ; 105: 106893, 2020 Sep.
Article En | MEDLINE | ID: mdl-32619502

INTRODUCTION: Drug-induced inotropic change is a risk factor in drug development; thus, de-risking is desired in the early stages of drug development. Unlike proarrhythmic risk assessment using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), few in vitro models were validated to predict cardiac contractility. Motion field imaging (MFI), a high-resolution block matching-based optical flow technique, was expected to possess high quantitative predictivity in the detection of contraction speed. We aimed to establish an in vitro model to assess drug-induced contractile changes using hiPSC-CMs and MFI. METHODS: MFI was designed to noninvasively characterize cardiomyocyte contractile behavior by analyzing light microscope video images, and maximum contraction speed (MCS) was used as the index of contractility. Using MFI, 9 inactive compounds, 10 negative inotropes, and 10 positive inotropes were tested. Two negative chronotropes, ZD7288 and ivabradine, were also tested. To determine the sensitivity and specificity of the assay, the minimum effective concentration of the MCS was compared with the human effective total therapeutic concentration for 28 compounds in clinical use. RESULTS: For 8 negative and 8 positive inotropes, the effects observed in in vivo and clinical studies were detected in MFI assay. MFI assay showed negative chronotropic changes without inotropic changes. MFI assay presented sufficient specificity (83%) and sensitivity (88%), and RNA-sequence analysis provided an insight into the relationship between occurrence of the false compounds and target gene expression. DISCUSSION: We demonstrated the utility of MFI assay using hiPSC-CMs to assess drug-induced contractile function. These results will facilitate the de-risking of compounds during early drug development.


Cardiotonic Agents/adverse effects , Cardiotoxicity/diagnosis , Induced Pluripotent Stem Cells/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Cells, Cultured , Gene Expression/drug effects , Humans , Risk Factors , Sensitivity and Specificity , Video Recording/methods
14.
Curr Pharm Biotechnol ; 21(9): 807-828, 2020.
Article En | MEDLINE | ID: mdl-32321398

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.


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
15.
Toxicol Sci ; 173(2): 347-361, 2020 02 01.
Article En | MEDLINE | ID: mdl-31722436

Cholestasis resulting from hepatic bile acid efflux transporter inhibition may contribute to drug-induced liver injury (DILI). This condition is a common safety-related reason for drug attrition and withdrawal. To screen for safety risks associated with efflux transport inhibition, we developed a high-throughput cellular assay for different drug discovery phases. Hepatocytes isolated from chimeric mice with humanized livers presented gene expression resembling that of the human liver and demonstrated apical membrane polarity when sandwiched between Matrigel and collagen. The fluorescent bile acid-derivative cholyl-l-lysyl-fluorescein (CLF) was used to quantify drug-induced efflux transport inhibition in hepatocytes. Cyclosporine inhibited CLF accumulation in the apical bile canalicular lumen in a concentration-dependent manner. The assay had equivalent predictive power to a primary human hepatocyte-based assay and greater predictive power than an assay performed with rat hepatocytes. Predictive power was tested using 45 pharmaceutical compounds, and 91.3% of the compounds with cholestatic potential (21/23) had margins (IC50/Cmax) < 20. In contrast, 90.9% (20/22) of compounds without cholestatic potential had IC50/Cmax>20. Assay sensitivity and specificity were 91.3% and 90.9%, respectively. We suggest that this improved assay performance could result from higher expression of efflux transporters, metabolic pathways, and/or species differences. Given the long-term supply of cells from the same donor, the humanized mouse-derived hepatocyte-based CLF efflux assay could be a valuable tool for predicting cholestatic DILI.


ATP Binding Cassette Transporter, Subfamily B, Member 11/antagonists & inhibitors , Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , High-Throughput Screening Assays/methods , Animals , Bile Canaliculi/metabolism , Chemical and Drug Induced Liver Injury/genetics , Cyclosporine/pharmacology , Gene Expression , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Mice , Mice, Transgenic
16.
Toxicol Appl Pharmacol ; 383: 114761, 2019 11 15.
Article En | MEDLINE | ID: mdl-31533062

Recent developments of novel targeted therapies are contributing to the increased long-term survival of cancer patients; however, drug-induced cardiotoxicity induced by cancer drugs remains a serious problem in clinical settings. Nevertheless, there are few in vitro cell-based assays available to predict this toxicity, especially from the aspect of morphology. Here, we developed a simple two-dimensional (2D) morphological assessment system, 2DMA, to predict drug-induced cardiotoxicity in cancer patients using human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with image-based high-content analysis in a high-throughput manner. To assess the effects of drugs on cardiomyocytes, we treated iPSC-CMs with 28 marketed pharmaceuticals and measured two key parameters: number of cell nuclei and sarcomere morphology. Drugs that significantly perturbed these two parameters at concentrations ≤30 times the human Cmax value were regarded as positive in the test. Based on these criteria, the sensitivity and specificity of the 2DMA system were 81% and 100%, respectively. Moreover, the translational predictability of 2DMA was comparable with that of a three-dimensional cardiotoxicity assay. RNA sequencing further revealed that the expression levels of several genes related to sarcomere components decreased following treatment with sunitinib, suggesting that inhibition of the synthesis of proteins that comprise the sarcomere contributes to drug-induced sarcomere disruption. Based on these features, the 2DMA system provides mechanistic insight with high predictability of cancer drug-induced cardiotoxicity in humans, and could thus contribute to the reduction of drug attrition rates at early stages of drug development.


Antineoplastic Agents/toxicity , Cardiotoxins/toxicity , Induced Pluripotent Stem Cells/drug effects , Microscopy, Electron/methods , Myocytes, Cardiac/drug effects , Cardiotoxicity/pathology , Cell Culture Techniques/methods , Cells, Cultured , Fluorescent Dyes/analysis , Forecasting , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/pathology
17.
Cell Metab ; 30(2): 374-384.e6, 2019 08 06.
Article En | MEDLINE | ID: mdl-31155493

Human organoid systems recapitulate in vivo organ architecture yet fail to capture complex pathologies such as inflammation and fibrosis. Here, using 11 different healthy and diseased pluripotent stem cell lines, we developed a reproducible method to derive multi-cellular human liver organoids composed of hepatocyte-, stellate-, and Kupffer-like cells that exhibit transcriptomic resemblance to in vivo-derived tissues. Under free fatty acid treatment, organoids, but not reaggregated cocultured spheroids, recapitulated key features of steatohepatitis, including steatosis, inflammation, and fibrosis phenotypes in a successive manner. Interestingly, an organoid-level biophysical readout with atomic force microscopy demonstrated that organoid stiffening reflects the fibrosis severity. Furthermore, organoids from patients with genetic dysfunction of lysosomal acid lipase phenocopied severe steatohepatitis, rescued by FXR agonism-mediated reactive oxygen species suppression. The presented key methodology and preliminary results offer a new approach for studying a personalized basis for inflammation and fibrosis in humans, thus facilitating the discovery of effective treatments.


Fatty Liver/pathology , Models, Biological , Organoids/cytology , Organoids/pathology , Pluripotent Stem Cells/cytology , Cells, Cultured , Fatty Liver/metabolism , Humans , Male
18.
J Stem Cells Regen Med ; 15(2): 45-51, 2019.
Article En | MEDLINE | ID: mdl-31983857

Human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) have attracted attention as a novel tool for drug safety screening and several differentiation protocols of hiPSC lines into cardiomyocytes have been reported; the standardization of these protocols will expand their applications for safety assessments such as "clinical safety trial-on-dish". Bone morphogenetic protein 4 (BMP4) is an important factor in promoting mesoderm differentiation and BMP4 treatment has been used at the early stage of cardiac differentiation into different hiPSCs. In the present study, we evaluated the effects of BMP4 treatment at the early stage of cardiac differentiation. We performed gene expression profiling of the germ layer during mesoderm differentiation of hiPSCs derived from three different donors. The expression of T (a mesoderm marker) and GATA6 (an endoderm marker) increased and that of PAX6 (a neuroectoderm marker) decreased in pooled embryoid bodies (EBs) after BMP4 treatment. Single-cell gene expression analysis revealed that mesodermal and mesendodermal populations increased in EBs derived from 253G1. Finally, BMP4 treatment increased mesodermal and mesendodermal populations compared with that without BMP4 in two other hiPSC lines, confirming the reproducibility of multiple hiPSC lines. Thus, our results suggest that BMP4 treatment increases mesodermal and mesendodermal populations at the early stage of cardiac differentiation in different hiPSC lines.

19.
Regul Toxicol Pharmacol ; 88: 238-251, 2017 Aug.
Article En | MEDLINE | ID: mdl-28634147

With the aim of reconsidering ICH S7B and E14 guidelines, a new in vitro assay system has been subjected to worldwide validation to establish a better prediction platform for potential drug-induced QT prolongation and the consequent TdP in clinical practice. In Japan, CSAHi HEART team has been working on hiPS-CMs in the MEA (hiPS-CMs/MEA) under a standardized protocol and found no inter-facility or lot-to-lot variability for proarrhythmic risk assessment of 7 reference compounds. In this study, we evaluated the responses of hiPS-CMs/MEA to another 31 reference compounds associated with cardiac toxicities, and gene expression to further clarify the electrophysiological characteristics over the course of culture period. The hiPS-CMs/MEA assay accurately predicted reference compounds potential for arrhythmogenesis, and yielded results that showed better correlation with target concentrations of QTc prolongation or TdP in clinical setting than other current in vitro and in vivo assays. Gene expression analyses revealed consistent profiles in all samples within and among the testing facilities. This report would provide CiPA with informative guidance on the use of the hiPS-CMs/MEA assay, and promote the establishment of a new paradigm, beyond conventional in vitro and in vivo assays for cardiac safety assessment of new drugs.


Long QT Syndrome/chemically induced , Myocytes, Cardiac/drug effects , Arrhythmias, Cardiac/chemically induced , Cardiotonic Agents/toxicity , Electrodes , Gene Expression , Guidelines as Topic , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Ion Channel Gating/genetics , Japan , Myocardial Contraction/genetics , Myocytes, Cardiac/physiology
20.
Stem Cell Reports ; 8(2): 226-234, 2017 02 14.
Article En | MEDLINE | ID: mdl-28111276

To predict drug-induced serious adverse events (SAE) in clinical trials, a model using a panel of cells derived from human induced pluripotent stem cells (hiPSCs) of individuals with different susceptibilities could facilitate major advancements in translational research in terms of safety and pharmaco-economics. However, it is unclear whether hiPSC-derived cells can recapitulate interindividual differences in drug-induced SAE susceptibility in populations not having genetic disorders such as healthy subjects. Here, we evaluated individual differences in SAE susceptibility based on an in vitro model using hiPSC-derived cardiomyocytes (hiPSC-CMs) as a pilot study. hiPSCs were generated from blood samples of ten healthy volunteers with different susceptibilities to moxifloxacin (Mox)-induced QT prolongation. Different Mox-induced field potential duration (FPD) prolongation values were observed in the hiPSC-CMs from each individual. Interestingly, the QT interval was significantly positively correlated with FPD at clinically relevant concentrations (r > 0.66) in multiple analyses including concentration-QT analysis. Genomic analysis showed no interindividual significant differences in known target-binding sites for Mox and other drugs such as the hERG channel subunit, and baseline QT ranges were normal. The results suggest that hiPSC-CMs from healthy subjects recapitulate susceptibility to Mox-induced QT prolongation and provide proof of concept for in vitro preclinical trials.


Electrophysiological Phenomena/drug effects , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Action Potentials/drug effects , Alleles , Cell Differentiation , ERG1 Potassium Channel/genetics , ERG1 Potassium Channel/metabolism , Electrocardiography , Gene Expression Profiling , Gene Frequency , Healthy Volunteers , Heart Conduction System/drug effects , Humans , Male , Mutation , Myocytes, Cardiac/cytology , Polymorphism, Single Nucleotide
...