Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Toxicol Appl Pharmacol ; 394: 114961, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32209365

RESUMO

INTRODUCTION: hERG block potency is widely used to calculate a drug's safety margin against its torsadogenic potential. Previous studies are confounded by use of different patch clamp electrophysiology protocols and a lack of statistical quantification of experimental variability. Since the new cardiac safety paradigm being discussed by the International Council for Harmonisation promotes a tighter integration of nonclinical and clinical data for torsadogenic risk assessment, a more systematic approach to estimate the hERG block potency and safety margin is needed. METHODS: A cross-industry study was performed to collect hERG data on 28 drugs with known torsadogenic risk using a standardized experimental protocol. A Bayesian hierarchical modeling (BHM) approach was used to assess the hERG block potency of these drugs by quantifying both the inter-site and intra-site variability. A modeling and simulation study was also done to evaluate protocol-dependent changes in hERG potency estimates. RESULTS: A systematic approach to estimate hERG block potency is established. The impact of choosing a safety margin threshold on torsadogenic risk evaluation is explored based on the posterior distributions of hERG potency estimated by this method. The modeling and simulation results suggest any potency estimate is specific to the protocol used. DISCUSSION: This methodology can estimate hERG block potency specific to a given voltage protocol. The relationship between safety margin thresholds and torsadogenic risk predictivity suggests the threshold should be tailored to each specific context of use, and safety margin evaluation may need to be integrated with other information to form a more comprehensive risk assessment.


Assuntos
Canal de Potássio ERG1/antagonistas & inibidores , Medição de Risco/métodos , Torsades de Pointes/induzido quimicamente , Teorema de Bayes , Simulação por Computador , Humanos , Modelos Biológicos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Segurança , Torsades de Pointes/fisiopatologia
2.
J Neurosci ; 36(16): 4408-14, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-27098685

RESUMO

α-Synuclein (α-syn) missense and multiplication mutations have been suggested to cause neurodegenerative diseases, including Parkinson's disease (PD) and dementia with Lewy bodies. Before causing the progressive neuronal loss, α-syn mutations impair exocytosis, which may contribute to eventual neurodegeneration. To understand how α-syn mutations impair exocytosis, we developed a mouse model that selectively expressed PD-related human α-syn A53T (h-α-synA53T) mutation at the calyx of Held terminals, where release mechanisms can be dissected with a patch-clamping technique. With capacitance measurement of endocytosis, we reported that h-α-synA53T, either expressed transgenically or dialyzed in the short term in calyces, inhibited two of the most common forms of endocytosis, the slow and rapid vesicle endocytosis at mammalian central synapses. The expression of h-α-synA53Tin calyces also inhibited vesicle replenishment to the readily releasable pool. These findings may help to understand how α-syn mutations impair neurotransmission before neurodegeneration. SIGNIFICANCE STATEMENT: α-Synuclein (α-syn) missense or multiplication mutations may cause neurodegenerative diseases, such as Parkinson's disease and dementia with Lewy bodies. The initial impact of α-syn mutations before neuronal loss is impairment of exocytosis, which may contribute to eventual neurodegeneration. The mechanism underlying impairment of exocytosis is poorly understood. Here we report that an α-syn mutant, the human α-syn A53T, inhibited two of the most commonly observed forms of endocytosis, slow and rapid endocytosis, at a mammalian central synapse. We also found that α-syn A53T inhibited vesicle replenishment to the readily releasable pool. These results may contribute to accounting for the widely observed early synaptic impairment caused by α-syn mutations in the progression toward neurodegeneration.


Assuntos
Endocitose/genética , Mutação/genética , Terminações Nervosas/fisiologia , Terminações Pré-Sinápticas/fisiologia , alfa-Sinucleína/genética , Animais , Tronco Encefálico/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , alfa-Sinucleína/metabolismo
4.
J Neurosci ; 33(20): 8820-6, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23678124

RESUMO

Studies over the last decade using FM dyes to label vesicles at many terminals, including the calyx-type nerve terminal, led to a well accepted "principle" that only a small fraction of vesicles (∼5-20%) participate in recycling under physiological conditions. This principle imposes a large challenge in maintaining synaptic transmission during repetitive firing, because the small recycling pool may limit the number of available vesicles for release and nerve terminals would have to distinguish the recycling pool from the reserve pool and keep reserve pool vesicles from being used. By recording the presynaptic capacitance changes and the postsynaptic EPSC at rat calyx of Held synapses in the absence or presence of transmitter glutamate in nerve terminals, we developed a new method to count functional recycling vesicles. We found that essentially all vesicles in calyces participated in recycling, challenging the small-recycling-pool principle established by FM dye labeling. Nerve terminals may use all available vesicles to maximize their ability in maintaining synaptic transmission during repetitive firing.


Assuntos
Endocitose/fisiologia , Terminações Pré-Sinápticas/fisiologia , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/fisiologia , Animais , Animais Recém-Nascidos , Biofísica , Tronco Encefálico/citologia , Estimulação Elétrica , Endocitose/efeitos dos fármacos , Inibidores Enzimáticos , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Feminino , Ácido Glutâmico/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Técnicas In Vitro , Ácido Cinurênico/farmacologia , Macrolídeos/farmacologia , Masculino , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Ratos , Ratos Wistar , Vesículas Sinápticas/efeitos dos fármacos
5.
Environ Technol ; : 1-10, 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38329084

RESUMO

Well-dispersed MIL-100(Fe) nanoparticles were synthesized under mild conditions and used to construct a photo-Fenton system (VMH system) with the assistance of visible-light irradiation and hydrogen peroxide. In such a VMH system, the MIL-100(Fe) has a high specific surface area and provides numerous Fe3+ active sites, thus accelerating the reaction of Fe3+ with photo-generated electrons under visible-light irradiation and generates Fe2+, and then the acquired Fe2+ can activate H2O2 to generate ⋅OH, accompanying with the oxidation of Fe2+ to Fe3+. Hence, the in-situ recycling of Fe2+/Fe3+ promotes the generation of ·OH, thus making the VMH system exhibits promising photocatalytic activity. The removal rate of ciprofloxacin in the VMH system is as high as 95.2% within 120 min photo-Fenton reaction, which is about 26 times higher than that of the Visible light/MIL-100(Fe) system. Moreover, the VMH system also exhibits strong degradation ability to other typical antibiotics, such as tetracycline, norfloxacin and cephalexin, and maintains high cyclic stability, revealing great practical application potential in the purification of antibiotic wastewater.

6.
Cell Discov ; 10(1): 62, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38862506

RESUMO

Membrane budding, which underlies fundamental processes like endocytosis, intracellular trafficking, and viral infection, is thought to involve membrane coat-forming proteins, including the most observed clathrin, to form Ω-shape profiles and helix-forming proteins like dynamin to constrict Ω-profiles' pores and thus mediate fission. Challenging this fundamental concept, we report that polymerized clathrin is required for Ω-profiles' pore closure and that clathrin around Ω-profiles' base/pore region mediates pore constriction/closure in neuroendocrine chromaffin cells. Mathematical modeling suggests that clathrin polymerization at Ω-profiles' base/pore region generates forces from its intrinsically curved shape to constrict/close the pore. This new fission function may exert broader impacts than clathrin's well-known coat-forming function during clathrin (coat)-dependent endocytosis, because it underlies not only clathrin (coat)-dependent endocytosis, but also diverse endocytic modes, including ultrafast, fast, slow, bulk, and overshoot endocytosis previously considered clathrin (coat)-independent in chromaffin cells. It mediates kiss-and-run fusion (fusion pore closure) previously considered bona fide clathrin-independent, and limits the vesicular content release rate. Furthermore, analogous to results in chromaffin cells, we found that clathrin is essential for fast and slow endocytosis at hippocampal synapses where clathrin was previously considered dispensable, suggesting clathrin in mediating synaptic vesicle endocytosis and fission. These results suggest that clathrin and likely other intrinsically curved coat proteins are a new class of fission proteins underlying vesicle budding and fusion. The half-a-century concept and studies that attribute vesicle-coat contents' function to Ω-profile formation and classify budding as coat-protein (e.g., clathrin)-dependent or -independent may need to be re-defined and re-examined by considering clathrin's pivotal role in pore constriction/closure.

7.
PLoS One ; 17(11): e0276995, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36417390

RESUMO

BACKGROUND AND PURPOSE: CaV1.2 channels contribute to action potential upstroke in pacemaker cells, plateau potential in working myocytes, and initiate excitation-contraction coupling. Understanding drug action on CaV1.2 channels may inform potential impact on cardiac function. However, literature shows large degrees of variability between CaV1.2 pharmacology generated by different laboratories, casting doubt regarding the utility of these data to predict or interpret clinical outcomes. This study examined experimental factors that may impact CaV1.2 pharmacology. EXPERIMENTAL APPROACH: Whole cell recordings were made on CaV1.2 overexpression cells. Current was evoked using a "step-step-ramp" waveform that elicited a step and a ramp current. Experimental factors examined were: 1) near physiological vs. room temperature for recording, 2) drug inhibition of the step vs. the ramp current, and 3) Ca2+ vs. Ba2+ as the charge carrier. Eight drugs were studied. KEY RESULTS: CaV1.2 current exhibited prominent rundown, exquisite temperature sensitivity, and required a high degree of series resistance compensation to optimize voltage control. Temperature-dependent effects were examined for verapamil and methadone. Verapamil's block potency shifted by up to 4X between room to near physiological temperature. Methadone exhibited facilitatory and inhibitory effects at near physiological temperature, and only inhibitory effect at room temperature. Most drugs inhibited the ramp current more potently than the step current-a preference enhanced when Ba2+ was the charge carrier. The slopes of the concentration-inhibition relationships for many drugs were shallow, temperature-dependent, and differed between the step and the ramp current. CONCLUSIONS AND IMPLICATIONS: All experimental factors examined affected CaV1.2 pharmacology. In addition, whole cell CaV1.2 current characteristics-rundown, temperature sensitivity, and impact of series resistance-are also factors that can impact pharmacology. Drug effects on CaV1.2 channels appear more complex than simple pore block mechanism. Normalizing laboratory-specific approaches is key to improve inter-laboratory data reproducibility. Releasing original electrophysiology records is essential to promote transparency and enable the independent evaluation of data quality.


Assuntos
Canais de Cálcio Tipo L , Excipientes , Canais de Cálcio Tipo L/fisiologia , Temperatura , Reprodutibilidade dos Testes , Verapamil/farmacologia , Metadona
8.
Nanoscale Adv ; 3(2): 347-352, 2021 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36131741

RESUMO

It is a great challenge to fabricate Janus inorganic/polymeric hybrid nanoparticles with both precisely controlled nanostructures and high yields. Herein, we report a new method to synthesize Janus Au@BCPs via UV light-initiated RAFT polymerization-induced self-assembly in situ at a high solid content. This strategy provides a promising alternative for achieving asymmetric hybrid nanoparticles with a controllable size, tunable morphology and convenient operation.

9.
Toxicol Sci ; 183(1): 214-226, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34240201

RESUMO

Bisphenol A (BPA) is a high-production volume chemical used to manufacture consumer and medical-grade plastic products. Due to its ubiquity, the general population can incur daily environmental exposure to BPA, whereas heightened exposure has been reported in intensive care patients and industrial workers. Due to health concerns, structural analogs are being explored as replacements for BPA. This study aimed to examine the direct effects of BPA on cardiac electrophysiology compared with recently developed alternatives, including BPS (bisphenol S) and BPF (bisphenol F). Whole-cell voltage-clamp recordings were performed on cell lines transfected to express the voltage-gated sodium channel (Nav1.5), L-type voltage-gated calcium channel (Cav1.2), or the rapidly activating delayed rectifier potassium channel (hERG). Cardiac electrophysiology parameters were measured using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) and intact, whole rat heart preparations. BPA was the most potent inhibitor of fast/peak (INa-P) and late (INa-L) sodium channel (IC50 = 55.3, 23.6 µM, respectively), L-type calcium channel (IC50 = 30.8 µM), and hERG channel current (IC50 = 127 µM). Inhibitory effects on L-type calcium channels were supported by microelectrode array recordings, which revealed a shortening of the extracellular field potential (akin to QT interval). BPA and BPF exposures slowed atrioventricular (AV) conduction and increased AV node refractoriness in isolated rat heart preparations, in a dose-dependent manner (BPA: +9.2% 0.001 µM, +95.7% 100 µM; BPF: +20.7% 100 µM). BPS did not alter any of the cardiac electrophysiology parameters tested. Results of this study demonstrate that BPA and BPF exert an immediate inhibitory effect on cardiac ion channels, whereas BPS is markedly less potent. Additional studies are necessary to fully elucidate the safety profile of bisphenol analogs on the heart.


Assuntos
Compostos Benzidrílicos , Técnicas Eletrofisiológicas Cardíacas , Animais , Compostos Benzidrílicos/toxicidade , Humanos , Fenóis , Ratos , Sulfonas
10.
Sci Rep ; 11(1): 8313, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33859333

RESUMO

The cation channel TRPML1 is an important regulator of lysosomal function and autophagy. Loss of TRPML1 is associated with neurodegeneration and lysosomal storage disease, while temporary inhibition of this ion channel has been proposed to be beneficial in cancer therapy. Currently available TRPML1 channel inhibitors are not TRPML isoform selective and block at least two of the three human isoforms. We have now identified the first highly potent and isoform-selective TRPML1 antagonist, the steroid 17ß-estradiol methyl ether (EDME). Two analogs of EDME, PRU-10 and PRU-12, characterized by their reduced activity at the estrogen receptor, have been identified through systematic chemical modification of the lead structure. EDME and its analogs, besides being promising new small molecule tool compounds for the investigation of TRPML1, selectively affect key features of TRPML1 function: autophagy induction and transcription factor EB (TFEB) translocation. In addition, they act as inhibitors of triple-negative breast cancer cell migration and invasion.


Assuntos
Autofagia/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Células Cultivadas , Feminino , Humanos , Invasividade Neoplásica , Neoplasias de Mama Triplo Negativas/patologia
11.
Polymers (Basel) ; 13(1)2020 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-33383843

RESUMO

In this research, we designed a feasible method to prepare composite films with high permittivity and significantly enhanced hydrophobic performance, which showed huge potential in the electrowetting field. TiO2 nanowire arrays were prepared by a one-step hydrothermal process, and poly(vinylidene fluoride-trifluoroethylene) (P(VDF-TrFE)) was spin-coated on the nanowire arrays to form composite, the surface of which was modified by electrospinning. Due to the great orientation of TiO2 nanowires, dipoles and space charges are in ordered arrangement along the electric field, and this strongly reinforced the Maxwell-Wagner-Sillars (MWS) polarization, thus the permittivity of the composite (TiO2 nanowire length/film thickness is 0.769) reaches 53 at 1 kHz, which is nearly 3 times higher than pure P(VDF-TrFE). Meanwhile the composite film possesses low dielectric loss (0.07) and low conductivity (2.69 × 10-9 S/cm), showing good insulation. The contact angle of the composite after electrospinning (about 137°) was greatly enhanced from pure P(VDF-TrFE) spin-coated film (about 89°), which can be attributed to the microrough structure built by P(VDF-TrFE) nanofibers.

12.
Materials (Basel) ; 13(10)2020 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-32443707

RESUMO

Influences and mechanisms of chemically synthesized nano-C-S-H gel addition on fresh properties of the cement-based materials with sucrose as a retarder were investigated in this study. The results showed that the flow value of the fresh cement paste was gradually but slightly reduced with increasing nano-C-S-H gel addition due to its fibrous but well-dispersed characteristic in both water and cement paste. The semi-adiabatic calorimetry testing results verified that incorporation of nano-C-S-H gel could greatly mitigate the retarding effect of sucrose on cement hydration. The total organic carbon (TOC) indicated that the addition of the nano-C-S-H gel helps to reduce adsorption of the sucrose molecules into the protective layer, thus the semi-permeability of the protective layer was less reduced and that is why the addition of the nano-C-S-H gel can mitigate the retardation caused by the sucrose. Through XRD analysis, it was found that the CH crystals are more prone to grow along the (0001) plane with larger size in the paste with nano-C-S-H addition before the induction period starts, because the C-S-H nanoparticles can form 3D network to slow down the diffusion rate of the released ions and eliminate the convection in the paste, thus suppress the 3D nucleation and growth of the CH crystals. The XRD analysis also indicated a refinement of the ettringite crystals in the paste with sucrose addition, but introduction of nano-C-S-H gel did not show further refinement, which was also verified by the SEM observation.

13.
PLoS One ; 15(11): e0241362, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33157550

RESUMO

Buprenorphine is a µ-opioid receptor (MOR) partial agonist used to manage pain and addiction. QTC prolongation that crosses the 10 msec threshold of regulatory concern was observed at a supratherapeutic dose in two thorough QT studies for the transdermal buprenorphine product BUTRANS®. Because QTC prolongation can be associated with Torsades de Pointes (TdP), a rare but potentially fatal ventricular arrhythmia, these results have led to further investigation of the electrophysiological effects of buprenorphine. Drug-induced QTC prolongation and TdP are most commonly caused by acute inhibition of hERG current (IhERG) that contribute to the repolarizing phase of the ventricular action potentials (APs). Concomitant inhibition of inward late Na+ (INaL) and/or L-type Ca2+ (ICaL) current can offer some protection against proarrhythmia. Therefore, we characterized the effects of buprenorphine and its major metabolite norbuprenorphine on cardiac hERG, Ca2+, and Na+ ion channels, as well as cardiac APs. For comparison, methadone, a MOR agonist associated with QTC prolongation and high TdP risk, and naltrexone and naloxone, two opioid receptor antagonists, were also studied. Whole cell recordings were performed at 37°C on cells stably expressing hERG, CaV1.2, and NaV1.5 proteins. Microelectrode array (MEA) recordings were made on human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). The results showed that buprenorphine, norbuprenorphine, naltrexone, and naloxone had no effect on IhERG, ICaL, INaL, and peak Na+ current (INaP) at clinically relevant concentrations. In contrast, methadone inhibited IhERG, ICaL, and INaL. Experiments on iPSC-CMs showed a lack of effect for buprenorphine, norbuprenorphine, naltrexone, and naloxone, and delayed repolarization for methadone at clinically relevant concentrations. The mechanism of QTC prolongation is opioid moiety-specific. This remains undefined for buprenorphine, while for methadone it involves direct hERG channel block. There is no evidence that buprenorphine use is associated with TdP. Whether this lack of TdP risk can be generalized to other drugs with QTC prolongation not mediated by acute hERG channel block warrants further study.


Assuntos
Buprenorfina/análogos & derivados , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Buprenorfina/farmacologia , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Metadona/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Naloxona/farmacologia , Naltrexona/farmacologia , Receptores Opioides/metabolismo , Fatores de Tempo
14.
J Pharmacol Toxicol Methods ; 105: 106890, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32574700

RESUMO

INTRODUCTION: In response to the ongoing shift of the regulatory cardiac safety paradigm, a recent White Paper proposed general principles for developing and implementing proarrhythmia risk prediction models. These principles included development strategies to validate models, and implementation strategies to ensure a model developed by one lab can be used by other labs in a consistent manner in the presence of lab-to-lab experimental variability. While the development strategies were illustrated through the validation of the model under the Comprehensive In vitro Proarrhythmia Assay (CiPA), the implementation strategies have not been adopted yet. METHODS: The proposed implementation strategies were applied to the CiPA model by performing a sensitivity analysis to identify a subset of calibration drugs that were most critical in determining the classification thresholds for proarrhythmia risk prediction. RESULTS: The selected calibration drugs were able to recapitulate classification thresholds close to those calculated from the full list of CiPA drugs. Using an illustrative dataset it was shown that a new lab could use these calibration drugs to establish its own classification thresholds (lab-specific calibration), and verify that the model prediction accuracy in the new lab is comparable to that in the original lab where the model was developed (lab-specific validation). DISCUSSION: This work used the CiPA model as an example to illustrate how to adopt the proposed model implementation strategies to select calibration drugs and perform lab-specific calibration and lab-specific validation. Generic in nature, these strategies could be generally applied to different proarrhythmia risk prediction models using various experimental systems under the new paradigm.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Bioensaio/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Preparações Farmacêuticas/administração & dosagem , Calibragem , Avaliação Pré-Clínica de Medicamentos/métodos , Eletrocardiografia/métodos , Humanos
15.
J Pharmacol Toxicol Methods ; 100: 106605, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31255744

RESUMO

INTRODUCTION: Cardiac late Na+ current (INaL) contributes to ventricular action potential duration. Pathological increase in INaL is arrhythmogenic, and inhibition of INaL offers protection against ventricular repolarization disturbance. Recently, two INaL datasets generated by different laboratories that assessed current inhibition by a panel of clinical drugs as a part of the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative were published. The results revealed a surprising degree of data variability despite of the use of a standardized voltage protocol. This study investigated whether remaining procedural differences related to experimental methods and data analysis associated with these datasets can produce differences in INaL pharmacology. METHODS: Whole cell voltage clamp recordings were performed on cells expressing NaV1.5 α- and ß1-subunits to study: 1) the impact of gating modifiers used to augment INaL (ATX-II vs. veratridine), internal solution composition (with vs. without ATP and GTP), and recording temperature (23 °C vs 37 °C) on stability of INaL measured across the duration of a patch clamp experiment; 2) mechanisms of each gating modifier on Na+ channels; and 3) effects of six drugs (lidocaine, mexiletine, chloroquine, ranolazine, ritonavir, and verapamil) on INaL induced by either gating modifier. RESULTS: Stability of INaL is affected by the choice of gating modifier, presence of nucleotides in the internal solution, and recording temperature. ATX-II and veratridine produced different changes in Na+ channel gating, inducing mechanistically distinct INaL. Drug potencies on inhibiting INaL were dependent on the choice of gating modifier and current region where drug effects were measured. DISCUSSION: INaL pharmacology can be impacted by all experimental factors examined in this study. The effect of gating modifier and current region used to quantify drug inhibition alone led to 30× difference in half inhibitory concentration (IC50) for ritonavir, demonstrating that substantial difference in drug inhibition can be produced. Drug potencies on inhibiting INaL derived from different patch clamp studies may thus not be generalizable. For INaL pharmacology to be useful for in silico modeling or interpreting drug-induced changes in cardiac action potentials or ECG, standardizing INaL experimental procedures including data analysis methods is necessary to minimize data variability.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Ventrículos do Coração/efeitos dos fármacos , Canais de Sódio/efeitos dos fármacos , Arritmias Cardíacas/diagnóstico , Simulação por Computador , Ventrículos do Coração/metabolismo , Humanos , Nucleotídeos/metabolismo , Técnicas de Patch-Clamp , Canais de Sódio/metabolismo , Temperatura
16.
Circ Arrhythm Electrophysiol ; 12(7): e007294, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31248280

RESUMO

BACKGROUND: Phthalates are used as plasticizers in the manufacturing of flexible, plastic medical products. Patients can be subjected to high phthalate exposure through contact with plastic medical devices. We aimed to investigate the cardiac safety and biocompatibility of mono-2-ethylhexyl phthalate (MEHP), a phthalate with documented exposure in intensive care patients. METHODS: Optical mapping of transmembrane voltage and pacing studies were performed on isolated, Langendorff-perfused rat hearts to assess cardiac electrophysiology after MEHP exposure compared with controls. MEHP dose was chosen based on reported blood concentrations after an exchange transfusion procedure. RESULTS: Thirty-minute exposure to MEHP increased the atrioventricular node (147 versus 107 ms) and ventricular (117 versus 77.5 ms) effective refractory periods, compared with controls. Optical mapping revealed prolonged action potential duration at slower pacing cycle lengths, akin to reverse use dependence. The plateau phase of the action potential duration restitution curve steepened and became monophasic in MEHP-exposed hearts (0.18 versus 0.06 slope). Action potential duration lengthening occurred during late-phase repolarization resulting in triangulation (70.3 versus 56.6 ms). MEHP exposure also slowed epicardial conduction velocity (35 versus 60 cm/s), which may be partly explained by inhibition of Nav1.5 (874 and 231 µmol/L half-maximal inhibitory concentration, fast and late sodium current). CONCLUSIONS: This study highlights the impact of acute MEHP exposure, using a clinically relevant dose, on cardiac electrophysiology in the intact heart. Heightened clinical exposure to plasticized medical products may have cardiac safety implications-given that action potential triangulation and electrical restitution modifications are a risk factor for early after depolarizations and cardiac arrhythmias.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Dietilexilftalato/análogos & derivados , Equipamentos e Provisões/efeitos adversos , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Plastificantes/toxicidade , Animais , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Simulação por Computador , Dietilexilftalato/toxicidade , Desenho de Equipamento , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Preparação de Coração Isolado , Masculino , Modelos Cardiovasculares , Ratos Sprague-Dawley , Período Refratário Eletrofisiológico/efeitos dos fármacos , Medição de Risco , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Fatores de Tempo , Imagens com Corantes Sensíveis à Voltagem
17.
Clin Pharmacol Ther ; 105(2): 466-475, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30151907

RESUMO

The International Council on Harmonization (ICH) S7B and E14 regulatory guidelines are sensitive but not specific for predicting which drugs are pro-arrhythmic. In response, the Comprehensive In Vitro Proarrhythmia Assay (CiPA) was proposed that integrates multi-ion channel pharmacology data in vitro into a human cardiomyocyte model in silico for proarrhythmia risk assessment. Previously, we reported the model optimization and proarrhythmia metric selection based on CiPA training drugs. In this study, we report the application of the prespecified model and metric to independent CiPA validation drugs. Over two validation datasets, the CiPA model performance meets all pre-specified measures for ranking and classifying validation drugs, and outperforms alternatives, despite some in vitro data differences between the two datasets due to different experimental conditions and quality control procedures. This suggests that the current CiPA model/metric may be fit for regulatory use, and standardization of experimental protocols and quality control criteria could increase the model prediction accuracy even further.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/epidemiologia , Simulação por Computador , Bases de Dados Factuais , Avaliação Pré-Clínica de Medicamentos/métodos , Canal de Potássio ERG1/efeitos dos fármacos , Humanos , Canais Iônicos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Reprodutibilidade dos Testes , Medição de Risco , Sensibilidade e Especificidade
18.
FASEB J ; 20(8): 1263-5, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16636101

RESUMO

Apomorphine (APO), a potent D1/D2 dopamine receptor agonist, is currently used as an antiparkinsonian drug. We have shown previously that APO stimulates synthesis and release of multiple trophic factors, such as brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF), in both mesencephalic and striatal neurons, thereby effectively preventing dopaminergic neuron loss in vitro. The present study was designed to investigate the effects of APO on fibroblast growth factor-2 (FGF-2) expression and regulation in astrocytes, and furthermore, to identify signaling mechanisms underlying these effects. Here, we show that FGF-2 expression is robustly induced in cultured astrocytes in response to APO. FGF-2 expression was proportional to APO concentration and time-dependent. Conversely, treatment with S-APO, a derivative of R-APO lacking DA receptor agonist activity, did not alter FGF-2 levels. APO treatment resulted in enhanced cytosol FGF-2 immunoreactivity, export of high MW forms of FGF-2 to the cytoplasm from the nucleus and increased extracellular release of FGF-2. Interestingly, both high and low MW forms of FGF-2 were detectable in conditioned medium of APO-treated cultures. This APO-induced effect was correlated with activation of D1 and D2 receptors, as it could be either mimicked by dopamine receptor agonists (SKF38393, quinpirole) or partially blocked by antagonists (SCH23390, SKF83566, haloperidol). Activation of the D1 receptor preferentially increased PKA activity, whereas activation of the D2 receptor only promoted phosphorylation of MAPK. Importantly, APO-modulated FGF-2 expression was independent of Akt/phosphoinositide 3-kinase signaling. These data suggest that APO can enhance biosynthesis and release of FGF-2 through activation of dopamine receptors in striatal astrocytes. Both cAMP/PKA and MEK/MAPK signaling cascades are major steps mediating this process.


Assuntos
Antiparkinsonianos/farmacologia , Apomorfina/farmacologia , Astrócitos/metabolismo , Agonistas de Dopamina/farmacologia , Fator 2 de Crescimento de Fibroblastos/biossíntese , Neurônios/citologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Sobrevivência Celular , Células Cultivadas , AMP Cíclico/metabolismo , Dopamina/metabolismo , Antagonistas dos Receptores de Dopamina D2 , Fator 2 de Crescimento de Fibroblastos/análise , Fator 2 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Neurônios/enzimologia , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D2/agonistas , Transdução de Sinais , Tirosina 3-Mono-Oxigenase/análise
19.
J Pharmacol Toxicol Methods ; 88(Pt 2): 109-122, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28830713

RESUMO

BACKGROUND: Loperamide (Immodium®) is indicated for symptomatic control of diarrhea. It is a µ-opioid receptor agonist, and recently has been associated with misuse and abuse. At therapeutic doses loperamide has not been associated with cardiotoxicity. However, loperamide overdose is associated with proarrhythmia and death - two effects that are likely attributable to its block of cardiac ion channels that are critical for generating action potentials. In this study, we defined loperamide-hERG channel interaction characteristics, and used a ventricular myocyte action potential model to compare loperamide's proarrhythmia propensity to twelve drugs with defined levels of clinical risk. METHODS AND RESULTS: Whole-cell voltage-clamp recordings were performed at 37°C on a HEK293 cell line stably expressing the hERG channel proteins, and loperamide was bath-applied to assess its effects on hERG current. Loperamide suppressed hERG current in a use- and voltage-dependent but frequency-independent manner, with a half-maximal inhibitory concentration <90nM. The onset of current suppression was concentration-dependent and appeared to follow a first-order reaction. Loperamide also altered the voltage-dependence of steady state hERG current properties. Electrophysiological data were integrated into a myocyte model that simulated dynamic drug-hERG channel interaction to estimate Torsade de Pointes risk through comparisons with reference drugs with defined clinical risk. In the context of overdose that would result in loperamide levels far exceeding those produced by therapeutic doses, loperamide is placed in the high risk category, alongside quinidine, bepridil, dofetilide, and sotalol. CONCLUSIONS: The combined in vitro and in silico approach provides mechanistic insight regarding the potential for loperamide to generate cardiotoxicity in overdose situations. This strategy holds promise for improving cardiac safety assessment.


Assuntos
Arritmias Cardíacas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Loperamida/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia , Relação Dose-Resposta a Droga , Canais de Potássio Éter-A-Go-Go/fisiologia , Células HEK293 , Humanos , Miócitos Cardíacos/fisiologia , Temperatura
20.
Circ Arrhythm Electrophysiol ; 10(2): e004628, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28202629

RESUMO

BACKGROUND: The current proarrhythmia safety testing paradigm, although highly efficient in preventing new torsadogenic drugs from entering the market, has important limitations that can restrict the development and use of valuable new therapeutics. The CiPA (Comprehensive in vitro Proarrhythmia Assay) proposes to overcome these limitations by evaluating drug effects on multiple cardiac ion channels in vitro and using these data in a predictive in silico model of the adult human ventricular myocyte. A set of drugs with known clinical torsade de pointes risk was selected to develop and calibrate the in silico model. METHODS AND RESULTS: Manual patch-clamp data assessing drug effects on expressed cardiac ion channels were integrated into the O'Hara-Rudy myocyte model modified to include dynamic drug-hERG channel (human Ether-à-go-go-Related Gene) interactions. Together with multichannel pharmacology data, this model predicts that compounds with high torsadogenic risk are more likely to be trapped within the hERG channel and show stronger reverse use dependency of action potential prolongation. Furthermore, drug-induced changes in the amount of electronic charge carried by the late sodium and L-type calcium currents was evaluated as a potential metric for assigning torsadogenic risk. CONCLUSIONS: Modeling dynamic drug-hERG channel interactions and multi-ion channel pharmacology improves the prediction of torsadogenic risk. With further development, these methods have the potential to improve the regulatory assessment of drug safety models under the CiPA paradigm.


Assuntos
Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Síndrome do QT Longo/induzido quimicamente , Torsades de Pointes/induzido quimicamente , Biomarcadores/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Canais Iônicos/efeitos dos fármacos , Cinética , Síndrome do QT Longo/fisiopatologia , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Medição de Risco , Torsades de Pointes/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA