Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
J Pharmacol Sci ; 153(3): 153-160, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37770156

RESUMO

Transporter-mediated clearance is determined by two factors, its single-molecule clearance, and expression level. However, no reliable method has been developed to evaluate them separately. This study aimed to develop a reliable method for evaluating the single-molecule activity of membrane transporters, such as organic anion transporting polypeptide (OATP) 2B1. HEK293 cells that co-expressed large conductance calcium-activated potassium (BK) channel and OATP2B1 were established and used for the following experiments. i) BK channel-mediated whole-cell conductance was measured using patch-clamp technique and divided by its unitary conductance to estimate the number of channels on plasma membrane (QI). ii) Using plasma membrane fraction, quantitative targeted absolute proteomics determined the stoichiometric ratio (ρ) of OATP2B1 to BK channel. iii) The uptake of estrone 3-sulfate was evaluated to calculate the Michaelis constant and uptake clearance (CL) per cell. Single-molecule clearance (CLint) was calculated by dividing CL by QI·ρ. QI and ρ values were estimated to be 916 and 2.16, respectively, yielding CLint of 5.23 fL/min/molecule. We successfully developed a novel method to reliably measure the single-molecule activity of a transporter, which could be used to evaluate the influences of factors such as genetic variations and post-translational modifications on the intrinsic activity of transporters.

2.
J Pharmacol Exp Ther ; 376(3): 454-462, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33376149

RESUMO

The activation of potassium channels and the ensuing hyperpolarization in skeletal myoblasts are essential for myogenic differentiation. However, the effects of K+ channel opening in myoblasts on skeletal muscle mass are unclear. Our previous study revealed that pharmacological activation of intermediate conductance Ca2+-activated K+ channels (IKCa channels) increases myotube formation. In this study, we investigated the effects of 5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one (DCEBIO), a Ca2+-activated K+ channel opener, on the mass of skeletal muscle. Application of DCEBIO to C2C12 cells during myogenesis increased the diameter of C2C12 myotubes in a concentration-dependent manner. This DCEBIO-induced hypertrophy was abolished by gene silencing of IKCa channels. However, it was resistant to 1 µM but sensitive to 10 µM TRAM-34, a specific IKCa channel blocker. Furthermore, DCEBIO reduced the mitochondrial membrane potential by opening IKCa channels. Therefore, DCEBIO should increase myotube mass by opening of IKCa channels distributed in mitochondria. Pharmacological studies revealed that mitochondrial reactive oxygen species (mitoROS), Akt, and mammalian target of rapamycin (mTOR) are involved in DCEBIO-induced myotube hypertrophy. An additional study demonstrated that DCEBIO-induced muscle hypertrophic effects are only observed when applied in the early stage of myogenic differentiation. In an in vitro myotube inflammatory atrophy experiment, DCEBIO attenuated the reduction of myotube diameter induced by endotoxin. Thus, we concluded that DCEBIO increases muscle mass by activating the IKCa channel/mitoROS/Akt/mTOR pathway. Our study suggests the potential of DCEBIO in the treatment of muscle wasting diseases. SIGNIFICANCE STATEMENT: Our study shows that 5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one (DCEBIO), a small molecule opener of Ca2+-activated K+ channel, increased muscle diameter via the mitochondrial reactive oxygen species/Akt/mammalian target of rapamycin pathway. And DCEBIO overwhelms C2C12 myotube atrophy induced by endotoxin challenge. Our report should inform novel role of K+ channel in muscle development and novel usage of K+ channel opener such as for the treatment of muscle wasting diseases.


Assuntos
Benzimidazóis/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/citologia , Canais de Potássio Cálcio-Ativados/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Canais de Potássio Cálcio-Ativados/química , Transdução de Sinais/efeitos dos fármacos
3.
PLoS Comput Biol ; 16(8): e1008109, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32797034

RESUMO

In the last decade, there has been tremendous progress in identifying genetic anomalies linked to clinical disease. New experimental platforms have connected genetic variants to mechanisms underlying disruption of cellular and organ behavior and the emergence of proarrhythmic cardiac phenotypes. The development of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) signifies an important advance in the study of genetic disease in a patient-specific context. However, considerable limitations of iPSC-CM technologies have not been addressed: 1) phenotypic variability in apparently identical genotype perturbations, 2) low-throughput electrophysiological measurements, and 3) an immature phenotype which may impact translation to adult cardiac response. We have developed a computational approach intended to address these problems. We applied our recent iPSC-CM computational model to predict the proarrhythmic risk of 40 KCNQ1 genetic variants. An IKs computational model was fit to experimental data for each mutation, and the impact of each mutation was simulated in a population of iPSC-CM models. Using a test set of 15 KCNQ1 mutations with known clinical long QT phenotypes, we developed a method to stratify the effects of KCNQ1 mutations based on proarrhythmic markers. We utilized this method to predict the severity of the remaining 25 KCNQ1 mutations with unknown clinical significance. Tremendous phenotypic variability was observed in the iPSC-CM model population following mutant perturbations. A key novelty is our reporting of the impact of individual KCNQ1 mutant models on adult ventricular cardiomyocyte electrophysiology, allowing for prediction of mutant impact across the continuum of aging. This serves as a first step toward translating predicted response in the iPSC-CM model to predicted response of the adult ventricular myocyte given the same genetic mutation. As a whole, this study presents a new computational framework that serves as a high throughput method to evaluate risk of genetic mutations based-on proarrhythmic behavior in phenotypically variable populations.


Assuntos
Canal de Potássio KCNQ1/genética , Modelos Cardiovasculares , Mutação/genética , Miócitos Cardíacos , Arritmias Cardíacas/genética , Biologia Computacional , Predisposição Genética para Doença/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/classificação , Miócitos Cardíacos/citologia
4.
J Physiol ; 597(17): 4533-4564, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31278749

RESUMO

KEY POINTS: Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) capture patient-specific genotype-phenotype relationships, as well as cell-to-cell variability of cardiac electrical activity Computational modelling and simulation provide a high throughput approach to reconcile multiple datasets describing physiological variability, and also identify vulnerable parameter regimes We have developed a whole-cell model of iPSC-CMs, composed of single exponential voltage-dependent gating variable rate constants, parameterized to fit experimental iPSC-CM outputs We have utilized experimental data across multiple laboratories to model experimental variability and investigate subcellular phenotypic mechanisms in iPSC-CMs This framework links molecular mechanisms to cellular-level outputs by revealing unique subsets of model parameters linked to known iPSC-CM phenotypes ABSTRACT: There is a profound need to develop a strategy for predicting patient-to-patient vulnerability in the emergence of cardiac arrhythmia. A promising in vitro method to address patient-specific proclivity to cardiac disease utilizes induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). A major strength of this approach is that iPSC-CMs contain donor genetic information and therefore capture patient-specific genotype-phenotype relationships. A cited detriment of iPSC-CMs is the cell-to-cell variability observed in electrical activity. We postulated, however, that cell-to-cell variability may constitute a strength when appropriately utilized in a computational framework to build cell populations that can be employed to identify phenotypic mechanisms and pinpoint key sensitive parameters. Thus, we have exploited variation in experimental data across multiple laboratories to develop a computational framework for investigating subcellular phenotypic mechanisms. We have developed a whole-cell model of iPSC-CMs composed of simple model components comprising ion channel models with single exponential voltage-dependent gating variable rate constants, parameterized to fit experimental iPSC-CM data for all major ionic currents. By optimizing ionic current model parameters to multiple experimental datasets, we incorporate experimentally-observed variability in the ionic currents. The resulting population of cellular models predicts robust inter-subject variability in iPSC-CMs. This approach links molecular mechanisms to known cellular-level iPSC-CM phenotypes, as shown by comparing immature and mature subpopulations of models to analyse the contributing factors underlying each phenotype. In the future, the presented models can be readily expanded to include genetic mutations and pharmacological interventions for studying the mechanisms of rare events, such as arrhythmia triggers.


Assuntos
Arritmias Cardíacas/fisiopatologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/fisiologia , Potenciais de Ação/fisiologia , Doença do Sistema de Condução Cardíaco/fisiopatologia , Simulação por Computador , Humanos , Armazenamento e Recuperação da Informação , Fenótipo
5.
J Pharmacol Sci ; 139(4): 259-265, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30962088

RESUMO

Sex hormones, such as testosterone, progesterone, and 17ß-estradiol, control various physiological functions. This review focuses on the sex hormonal regulation of K+ channels and the effects of such regulation on electrophysiological and contractile functions of muscles. In the cardiac tissue, testosterone and progesterone shorten action potential, and estrogen lengthens QT interval, a marker of increased risk of ventricular tachyarrhythmias. We have shown that testosterone and progesterone in physiological concentration activate KCNQ1 channels via membrane-delimited sex hormone receptor/eNOS pathways to shorten the action potential duration. Mitochondrial K+ channels are also involved in the protection of cardiac muscle. Testosterone and 17ß-estradiol directly activate mitochondrial inner membrane K+ channels (Ca2+ activated K+ channel (KCa channel) and ATP-sensitive K+ channel (KATP channel)) that are involved in ischemic preconditioning and cardiac protection. During pregnancy, uterine blood flow increases to support fetal growth and development. It has been reported that 17ß-estradiol directly activates large-conductance Ca2+-activated K+ channel (BKCa channel) attenuating arterial contraction. Furthermore, 17ß-estradiol increases expression of BKCa channel ß1 subunit which enhances BKCa channel activity by DNA demethylation. These findings are useful for understanding the mechanisms of sex or generation-dependent differences in the physiological and pathological functions of muscles, and the mechanisms of drug actions.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Estradiol/fisiologia , Contração Muscular/fisiologia , Músculos/metabolismo , Músculos/fisiologia , Canais de Potássio/metabolismo , Canais de Potássio/fisiologia , Progesterona/fisiologia , Testosterona/fisiologia , Animais , Humanos
6.
J Pharmacol Sci ; 140(4): 345-349, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31521491

RESUMO

Although the cardiotoxicity of anti-cancer drugs is an important issue, the underlying mechanisms remain unknown. To develop a sensitive assay system for cardiotoxicity, we examined effects of anti-cancer drugs on contractile functions of human iPS cell-derived cardiomyocytes by using non-invasive motion field imaging analysis with extended drug exposure time. We succeeded in continuously measuring stable contractile function. The continued exposure revealed that the difference in cardiotoxicity between cardiotoxic doxorubicin and less toxic erlotinib was more evident after 8 days of treatment than with 3 days of treatment, suggesting that continued exposure improved the predictive power for cardiotoxicity of anti-cancer drugs.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Cardiotoxicidade/etiologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Células Cultivadas , Doxorrubicina/efeitos adversos , Doxorrubicina/farmacologia , Cloridrato de Erlotinib/efeitos adversos , Cloridrato de Erlotinib/farmacologia , Humanos , Contração Miocárdica/efeitos dos fármacos
7.
J Pharmacol Sci ; 140(4): 325-330, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31279582

RESUMO

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are a valuable tool to characterize the pharmacology and toxic effects of drugs on heart cells. In particular, hiPSC-CMs can be used to identify drugs that generate arrhythmias. However, it is unclear whether the expression of genes related to generation of CM action potentials differs between hiPSC-CM cell lines and the mature human heart. To address this, we obtained accurate gene expression profiles of commercially available hiPSC-CM cell lines with quantitative real time RT-PCR analysis. Expression analysis of ten cardiac proteins important for generation of action potentials and three cardiac proteins important for muscle contractility was performed using GAPDH for normalization. Comparison revealed large variations in expression levels among hiPSC-CM cell lines and between hiPSC-CMs and normal human heart. In general, gene expression in hiPSC-CM cell lines was more similar to an immature, stem-like cell than a mature cardiomyocyte from human heart samples. These results provide quantitative information about differences in gene expression between hiPSC-CM cell lines, essential for interpreting pharmacology experiments. Our approach can be used as an experimental guideline for future research on gene expression in hiPSC-CMs.


Assuntos
Potenciais de Ação/genética , Expressão Gênica/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/fisiologia , Adulto , Arritmias Cardíacas/genética , Linhagem Celular , Coração/fisiologia , Humanos , Masculino , Contração Muscular/genética
8.
Proc Natl Acad Sci U S A ; 113(3): 775-80, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26739563

RESUMO

Heterodimeric amino acid transporters play crucial roles in epithelial transport, as well as in cellular nutrition. Among them, the heterodimer of a membrane protein b(0,+)AT/SLC7A9 and its auxiliary subunit rBAT/SLC3A1 is responsible for cystine reabsorption in renal proximal tubules. The mutations in either subunit cause cystinuria, an inherited amino aciduria with impaired renal reabsorption of cystine and dibasic amino acids. However, an unsolved paradox is that rBAT is highly expressed in the S3 segment, the late proximal tubules, whereas b(0,+)AT expression is highest in the S1 segment, the early proximal tubules, so that the presence of an unknown partner of rBAT in the S3 segment has been proposed. In this study, by means of coimmunoprecipitation followed by mass spectrometry, we have found that a membrane protein AGT1/SLC7A13 is the second partner of rBAT. AGT1 is localized in the apical membrane of the S3 segment, where it forms a heterodimer with rBAT. Depletion of rBAT in mice eliminates the expression of AGT1 in the renal apical membrane. We have reconstituted the purified AGT1-rBAT heterodimer into proteoliposomes and showed that AGT1 transports cystine, aspartate, and glutamate. In the apical membrane of the S3 segment, AGT1 is suggested to locate itself in close proximity to sodium-dependent acidic amino acid transporter EAAC1 for efficient functional coupling. EAAC1 is proposed to take up aspartate and glutamate released into luminal fluid by AGT1 due to its countertransport so that preventing the urinary loss of aspartate and glutamate. Taken all together, AGT1 is the long-postulated second cystine transporter in the S3 segment of proximal tubules and a possible candidate to be involved in isolated cystinuria.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Membrana Celular/metabolismo , Cistinúria/metabolismo , Túbulos Renais Proximais/metabolismo , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/genética , Animais , Anticorpos/metabolismo , Western Blotting , Transportador 3 de Aminoácido Excitatório/metabolismo , Feminino , Células HEK293 , Humanos , Imuno-Histoquímica , Hibridização In Situ , Rim/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Ligação Proteica , Multimerização Proteica , Proteolipídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Elementos de Resposta/genética
9.
J Physiol ; 595(14): 4695-4723, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28516454

RESUMO

KEY POINTS: This study represents a first step toward predicting mechanisms of sex-based arrhythmias that may lead to important developments in risk stratification and may inform future drug design and screening. We undertook simulations to reveal the conditions (i.e. pacing, drugs, sympathetic stimulation) required for triggering and sustaining reentrant arrhythmias. Using the recently solved cryo-EM structure for the Eag-family channel as a template, we revealed potential interactions of oestrogen with the pore loop hERG mutation (G604S). Molecular models suggest that oestrogen and dofetilide blockade can concur simultaneously in the hERG channel pore. ABSTRACT: Female sex is a risk factor for inherited and acquired long-QT associated torsade de pointes (TdP) arrhythmias, and sympathetic discharge is a major factor in triggering TdP in female long-QT syndrome patients. We used a combined experimental and computational approach to predict 'the perfect storm' of hormone concentration, IKr block and sympathetic stimulation that induces arrhythmia in females with inherited and acquired long-QT. More specifically, we developed mathematical models of acquired and inherited long-QT syndrome in male and female ventricular human myocytes by combining effects of a hormone and a hERG blocker, dofetilide, or hERG mutations. These 'male' and 'female' model myocytes and tissues then were used to predict how various sex-based differences underlie arrhythmia risk in the setting of acute sympathetic nervous system discharge. The model predicted increased risk for arrhythmia in females when acute sympathetic nervous system discharge was applied in the settings of both inherited and acquired long-QT syndrome. Females were predicted to have protection from arrhythmia induction when progesterone is high. Males were protected by the presence of testosterone. Structural modelling points towards two plausible and distinct mechanisms of oestrogen action enhancing torsadogenic effects: oestradiol interaction with hERG mutations in the pore loop containing G604 or with common TdP-related blockers in the intra-cavity binding site. Our study presents findings that constitute the first evidence linking structure to function mechanisms underlying female dominance of arousal-induced arrhythmias.


Assuntos
Nível de Alerta/fisiologia , Arritmias Cardíacas/fisiopatologia , Modelos Biológicos , Agonistas Adrenérgicos beta/farmacologia , Animais , Antiarrítmicos/farmacologia , Estradiol/farmacologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Cobaias , Isoproterenol/farmacologia , Masculino , Simulação de Acoplamento Molecular , Miócitos Cardíacos/fisiologia , Fenetilaminas/farmacologia , Caracteres Sexuais , Sulfonamidas/farmacologia
10.
J Pharmacol Sci ; 134(2): 75-85, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28615142

RESUMO

Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes hold great potentials to predict pro-arrhythmic risks in preclinical cardiac safety screening, although the hiPSC cardiomyocytes exhibit rather immature functional and structural characteristics, including spontaneous activity. Our physiological characterization and mathematical simulation showed that low expression of the inward-rectifier potassium (IK1) channel is a determinant of spontaneous activity. To understand impact of the low IK1 expression on the pharmacological properties, we tested if transduction of hiPSC-derived cardiomyocytes with KCNJ2, which encodes the IK1 channel, alters pharmacological response to cardiac repolarization processes. The transduction of KCNJ2 resulted in quiescent hiPSC-derived cardiomyocytes, which need pacing to elicit action potentials. Significant prolongation of paced action potential duration in KCNJ2-transduced hiPSC-derived cardiomyocytes was stably measured at 0.1 µM E-4031, although the same concentration of E-4031 ablated firing of non-treated hiPSC-derived cardiomyocytes. These results in single cells were confirmed by mathematical simulations. Using the hiPSC-derived cardiac sheets with KCNJ2-transduction, we also investigated effects of a range of drugs on field potential duration recorded at 1 Hz. The KCNJ2 overexpression in hiPSC-derived cardiomyocytes may contribute to evaluate a part of QT-prolonging drugs at toxicological concentrations with high accuracy.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/metabolismo , Bloqueadores dos Canais de Potássio/efeitos adversos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Avaliação Pré-Clínica de Medicamentos/métodos , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Piperidinas/efeitos adversos , Piridinas/efeitos adversos
11.
J Mol Cell Cardiol ; 90: 38-46, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26654778

RESUMO

BACKGROUND: Lifestyle-related diseases, such as obesity and dyslipidemia are important risk factors for atrial fibrillation (AF). However, the underlying mechanism linking these diseases and AF has not been fully investigated. METHODS: Adult male mice were fed a high-fat diet (HFD) or vehicle (NC) for 2 months. Electrocardiography and in vivo electrophysiological study were performed. Mice were then sacrificed for quantification of mRNA, microRNA, and protein in atria, in addition to histological analysis. Conduction velocity (CV) in right atrium was measured by optical mapping in Langendorff perfused hearts. Cultured atrial cardiomyocytes were treated with palmitate with or without a specific microRNA inhibitor. Twelve hours after stimulation, cells were lysed, and subjected to analysis with qPCR and Western blotting. RESULTS: HFD mice showed prolonged P wave duration, increased inducibility of sustained atrial tachycardia, and reduced atrial CV than NC mice. HFD mice also showed increased expression in inflammatory cytokines, whereas fibrotic area and signals relating fibrosis were not changed. HFD mice demonstrated reduced expression of Cx40 in mRNA and protein levels, and its lateralized expression in atria. MicroRNA array analysis revealed that miR-27b expression was up-regulated in HFD mice, and luciferase assay confirmed the direct interaction between miR-27b and Cx40 3'UTR. In palmitate-stimulated atrial cardiomyocytes, miR-27b up-regulation and Cx40 down-regulation were observed, while expression of inflammatory cytokines was not altered. Inhibition of miR-27b with antisense oligonucleotides reversed the alteration caused by palmitate stimulation. CONCLUSION: HFD may increase the vulnerability to atrial arrhythmia by down-regulation of Cx40 via miR-27b, rather than fibrosis, which is independent of inflammation.


Assuntos
Arritmia Sinusal/genética , Síndrome de Brugada/genética , Conexinas/genética , Dieta Hiperlipídica/efeitos adversos , MicroRNAs/genética , Regiões 3' não Traduzidas , Animais , Arritmia Sinusal/etiologia , Arritmia Sinusal/metabolismo , Arritmia Sinusal/patologia , Síndrome de Brugada/etiologia , Síndrome de Brugada/metabolismo , Síndrome de Brugada/patologia , Doença do Sistema de Condução Cardíaco , Linhagem Celular , Conexinas/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Técnicas de Cultura de Órgãos , Ácido Palmítico/farmacologia , Transdução de Sinais , Proteína alfa-5 de Junções Comunicantes
12.
J Pharmacol Sci ; 131(2): 141-5, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27179939

RESUMO

We investigated electrophysiological properties of human induced-pluripotent-stem-cell-derived and embryonic-stem-cell-derived cardiomyocytes, and analyzed action potential parameters by plotting their frequency distributions. In the both cell lines, the distribution analysis revealed that histograms of maximum upstroke velocity showed two subpopulations with similar intersection values. Sub-populations with faster maximum upstroke velocity showed significant prolongation of action potential durations by application of E-4031, whereas others did not, which may be partly due to shallower maximum diastolic potentials. We described electrophysiological and pharmacological properties of stem-cell-derived cardiomyocytes in the respective sub-populations, which provides a way to characterize diverse electrical properties of stem-cell-derived cardiomyocytes systematically.


Assuntos
Potenciais de Ação/fisiologia , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/citologia , Linhagem Celular , Humanos , Técnicas de Patch-Clamp
13.
Int Heart J ; 57(3): 341-9, 2016 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-27170476

RESUMO

Genome-wide association study has identified that the genetic variations at NOS1AP (neuronal nitric oxide synthase-1 adaptor protein) were associated with QT interval and sudden cardiac death (SCD). However, the mechanism linking a genetic variant of NOS1AP and SCD is poorly understood. We used Nos1ap knockout mice (Nos1ap(-/-)) to determine the involvement of Nos1ap in SCD, paying special attention to oxidative stress.At baseline, a surface electrocardiogram (ECG) and ultrasound echocardiography (UCG) showed no difference between Nos1ap(-/-) and wild-type (WT) mice. Oxidative stress was induced by a single injection of doxorubicin (Dox, 25 mg/kg). After Dox injection, Nos1ap(-/-) showed significantly higher mortality than WT (93.3 versus 16.0% at day 14, P < 0.01). ECG showed significantly longer QTc in Nos1ap(-/-) than WT, and UCG revealed significant reduction of fractional shortening (%FS) only in Nos1ap(-/-) after Dox injection. Spontaneous ventricular tachyarrhythmias were documented by telemetry recording after Dox injection only in Nos1ap(-/-). Ex vivo optical mapping revealed that the action potential duration (APD)90 was prolonged at baseline in Nos1ap(-/-), and administration of Dox lengthened APD90 more in Nos1ap(-/-) than in WT. The expression of Bnp and the H2O2 level were higher in Nos1ap(-/-) after Dox injection. Nos1ap(-/-) showed a reduced amplitude of calcium transient in isolated cardiomyocytes after Dox injection. Administration of the antioxidant N-acetyl-L-cysteine significantly reduced mortality of Nos1ap(-/-) by Dox injection, accompanied by prevention of QT prolongation and a reduction in %FS.Although Nos1ap(-/-) mice have apparently normal hearts, oxidative stress evokes ventricular tachyarrhythmia and heart failure, which may cause sudden cardiac death.


Assuntos
Acetilcisteína/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Morte Súbita Cardíaca , Insuficiência Cardíaca , Estresse Oxidativo/efeitos dos fármacos , Taquicardia Ventricular , Animais , Antioxidantes/farmacologia , Morte Súbita Cardíaca/etiologia , Morte Súbita Cardíaca/prevenção & controle , Eletrocardiografia/métodos , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/prevenção & controle , Camundongos , Camundongos Knockout , Modelos Cardiovasculares , Polimorfismo de Nucleotídeo Único , Taquicardia Ventricular/complicações , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/prevenção & controle
14.
J Mol Cell Cardiol ; 77: 178-91, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25257913

RESUMO

In this study, we used high-speed video microscopy with motion vector analysis to investigate the contractile characteristics of hiPS-CM monolayer, in addition to further characterizing the motion with extracellular field potential (FP), traction force and the Ca(2+) transient. Results of our traction force microscopy demonstrated that the force development of hiPS-CMs correlated well with the cellular deformation detected by the video microscopy with motion vector analysis. In the presence of verapamil and isoproterenol, contractile motion of hiPS-CMs showed alteration in accordance with the changes in fluorescence peak of the Ca(2+) transient, i.e., upstroke, decay, amplitude and full-width at half-maximum. Simultaneously recorded hiPS-CM motion and FP showed that there was a linear correlation between changes in the motion and field potential duration in response to verapamil (30-150nM), isoproterenol (0.1-10µM) and E-4031 (10-50nM). In addition, tetrodotoxin (3-30µM)-induced delay of sodium current was corresponded with the delay of the contraction onset of hiPS-CMs. These results indicate that the electrophysiological and functional behaviors of hiPS-CMs are quantitatively reflected in the contractile motion detected by this image-based technique. In the presence of 100nM E-4031, the occurrence of early after-depolarization-like negative deflection in FP was also detected in the hiPS-CM motion as a characteristic two-step relaxation pattern. These findings offer insights into the interpretation of the motion kinetics of the hiPS-CMs, and are relevant for understanding electrical and mechanical relationship in hiPS-CMs.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Contração Miocárdica , Miócitos Cardíacos/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Fenômenos Biomecânicos , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Diferenciação Celular , Células Cultivadas , Técnicas Eletrofisiológicas Cardíacas , Humanos , Isoproterenol/farmacologia , Cinética , Microscopia de Vídeo , Miócitos Cardíacos/efeitos dos fármacos , Piperidinas/farmacologia , Piridinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Verapamil/farmacologia
15.
Nihon Yakurigaku Zasshi ; 159(2): 112-117, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38432919

RESUMO

While sepsis mortality is reducing in developed countries due to advances in intensive care medicine, morbidity is increasing due to aging and obesity. ICU-acquired weakness (ICU-AW) is a respiratory and limb muscle weakness experienced by many sepsis survivors and is present in 50-75% of sepsis patients. ICU-AW can persist for several years, making reintegration of sepsis survivors difficult and leading to a secondary decrease in long-term survival. Exposure of septic patients to multiple muscle-damaging factors during ICU admission, including hyperglycemia, immobility, mechanical ventilation, administration of muscle relaxants, and administration of steroidal anti-inflammatory drugs, may compound the hyper cytokine, hyper nitric oxide, and hyper oxidative conditions, leading to the development of ICU-AW. However, the pathogenesis of ICU-AW remains unclear, and the pathophysiology of ICU-AW awaits further elucidation to develop therapeutic strategies. Recent ICU-AW studies have also revealed that skeletal muscle itself is a key organ in the inflammatory response and metabolic abnormalities in sepsis. In this article, we review the pathophysiology of skeletal muscle in sepsis and international trends in the development of therapeutic agents based on our research results.


Assuntos
Músculo Esquelético , Sepse , Humanos , Envelhecimento , Citocinas , Óxido Nítrico
16.
Yakugaku Zasshi ; 144(3): 251-255, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38432933

RESUMO

Cardiotoxicity induced by anti-cancer drugs is a significant concern for patients undergoing cancer treatment. Some anti-cancer drugs can damage cardiac muscle cells directly or indirectly, potentially leading to severe heart failure. Various risk factors, including the type and dosage of chemotherapy agents as well as patient background, contribute to the development of cardiotoxicity. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), which enable patient-specific toxicity prediction, hold great promise in this regard. However, the practical implementation of hiPSC-CMs-based prediction of anti-cancer drug-induced cardiotoxicity still faces hurdles. One major challenge involves establishing and optimizing experimental systems for evaluating contractile dysfunction, the ultimate output of heart failure, using hiPSC-CMs. Such efforts are currently underway globally, focusing on tailoring functional evaluation systems to the characteristics of hiPSC-CMs. In this paper, we provide an overview of the contraction mechanisms of cardiac cells and introduce a method of measuring contraction that we have developed, and discuss the current status of contractile function evaluation methods using hiPSC-CMs.


Assuntos
Antineoplásicos , Insuficiência Cardíaca , Células-Tronco Pluripotentes Induzidas , Humanos , Miócitos Cardíacos , Cardiotoxicidade/etiologia
17.
J Pharmacol Toxicol Methods ; 128: 107530, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38917571

RESUMO

INTRODUCTION: Cardiac safety assessment, such as lethal arrhythmias and contractility dysfunction, is critical during drug development. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been shown to be useful in predicting drug-induced proarrhythmic risk through international validation studies. Although cardiac contractility is another key function, fit-for-purpose hiPSC-CMs in evaluating drug-induced contractile dysfunction remain poorly understood. In this study, we investigated whether alignment of hiPSC-CMs on nanopatterned culture plates can assess drug-induced contractile changes more efficiently than non-aligned monolayer culture. METHODS: Aligned hiPSC-CMs were obtained by culturing on 96-well culture plates with a ridge-groove-ridge nanopattern on the bottom surface, while non-aligned hiPSC-CMs were cultured on regular 96-well plates. Next-generation sequencing and qPCR experiments were performed for gene expression analysis. Contractility of the hiPSC-CMs was assessed using an imaging-based motion analysis system. RESULTS: When cultured on nanopatterned plates, hiPSC-CMs exhibited an aligned morphology and enhanced expression of genes encoding proteins that regulate contractility, including myosin heavy chain, calcium channel, and ryanodine receptor. Compared to cultures on regular plates, the aligned hiPSC-CMs also showed both enhanced contraction and relaxation velocity. In addition, the aligned hiPSC-CMs showed a more physiological response to positive and negative inotropic agents, such as isoproterenol and verapamil. DISCUSSION: Taken together, the aligned hiPSC-CMs exhibited enhanced structural and functional properties, leading to an improved capacity for contractility assessment compared to the non-aligned cells. These findings suggest that the aligned hiPSC-CMs can be used to evaluate drug-induced cardiac contractile changes.

18.
J Pharmacol Sci ; 121(1): 1-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23238536

RESUMO

In short QT syndrome, inherited gain-of-function mutations in the human ether a-gogo-related gene (hERG) K(+) channel have been associated with development of fatal arrhythmias. This implies that drugs that activate hERG as a side effect may likewise pose significant arrhythmia risk. hERG activators have been found to have diverse mechanisms of activation, which may reflect their distinct binding sites. Recently, the new hERG activator ICA-105574 was introduced, which disables inactivation of the hERG channel with very high potency. We explored characteristics of this new drug in several experimental models. Patch clamp experiments were used to verify activation of hERG channels by ICA-105574 in human embryonic kidney cells stably-expressing hERG channels. ICA-105574 significantly shortened QT and QTc intervals and monophasic action potential duration (MAP(90)) in Langendorff-perfused guinea-pig hearts. We also administered ICA-105574 to anesthetized dogs while recording ECG and drug plasma concentrations. ICA-105574 (10 mg/kg) significantly shortened QT and QTc intervals, with a free plasma concentration of approximately 1.7 µM at the point of maximal effect. Our data showed that unbound ICA-105574 caused QT shortening in dogs at concentrations comparable to the half maximal effective concentration (EC(50), 0.42 µM) of hERG activation in the patch clamp studies.


Assuntos
Benzamidas/farmacologia , Eletrocardiografia/efeitos dos fármacos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/metabolismo , Coração/efeitos dos fármacos , Animais , Células Cultivadas , Cães , Relação Dose-Resposta a Droga , Cobaias , Humanos , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp
19.
Biol Pharm Bull ; 36(1): 8-12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23302631

RESUMO

Gender differences play a major role in the manifestation of cardiovascular disease including cardiac arrhythmias. In particular, female sex is an independent risk factor for development of torsade de pointes (TdP) arrhythmias not only in congenital long QT syndromes but also in acquired long QT syndromes which occur as adverse effects of existing drugs. Recent clinical and experimental studies suggest that the gender differences may stem, at least in part, from gender differences in cardiac repolarization process, that is longer rate-corrected QT (QT(C)) interval in women than in men. In women, QT(C) interval and arrhythmic risks in TdP alter cyclically during menstrual cycle, suggesting a critical role of female sex hormones in cardiac repolarization process. These gender differences in fundamental cardiac electrophysiology result from variable ion channel expression and diverse sex hormonal regulation via long term genomic and acute non-genomic pathways, and sex differences in drug responses and metabolisms. In particular, non-genomic actions of testosterone and progesterone on cardiac ion channels likely to contribute to the gender differences in cardiac repolarization processes.


Assuntos
Hormônios Esteroides Gonadais/fisiologia , Coração/fisiologia , Animais , Humanos , Canais Iônicos/fisiologia , Síndrome do QT Longo/fisiopatologia
20.
Circ J ; 76(2): 293-4, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22185712

RESUMO

The results presented by Shinohara et al(1) suggests that both the membrane clock and the Ca(2+) clock contribute to sinus node automaticity. Which is dominant depends on the region within the sinus node and the condition of the SNS. When you change the way you look at things, the things you look at change. This new hypothesis is very attractive, because it combines the membrane and Ca(2+) clocks. In fact, supporting data have been reported by the same parties.(11,12) Supportive studies by others are warranted.


Assuntos
Relógios Biológicos/fisiologia , Cálcio/farmacocinética , Potássio/farmacocinética , Nó Sinoatrial/citologia , Nó Sinoatrial/fisiologia , Taquicardia Sinusal/fisiopatologia , Animais , Masculino
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa