Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Toxicol Sci ; 191(1): 47-60, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36226800

RESUMO

Determining the potential cardiotoxicity and pro-arrhythmic effects of drug candidates remains one of the most relevant issues in the drug development pipeline (DDP). New methods enabling to perform more representative preclinical in vitro studies by exploiting induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) are under investigation to increase the translational power of the outcomes. Here we present a pharmacological campaign conducted to evaluate the drug-induced QT alterations and arrhythmic events on uHeart, a 3D miniaturized in vitro model of human myocardium encompassing iPSC-CM and dermal fibroblasts embedded in fibrin. uHeart was mechanically trained resulting in synchronously beating cardiac microtissues in 1 week, characterized by a clear field potential (FP) signal that was recorded by means of an integrated electrical system. A drug screening protocol compliant with the new International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines was established and uHeart was employed for testing the effect of 11 compounds acting on single or multiple cardiac ion channels and well-known to elicit QT prolongation or arrhythmic events in clinics. The alterations of uHeart's electrophysiological parameters such as the beating period, the FP duration, the FP amplitude, and the detection of arrhythmic events prior and after drug administration at incremental doses were effectively analyzed through a custom-developed algorithm. Results demonstrated the ability of uHeart to successfully anticipate clinical outcome and to predict the QT prolongation with a sensitivity of 83.3%, a specificity of 100% and an accuracy of 91.6%. Cardiotoxic concentrations of drugs were notably detected in the range of the clinical highest blood drug concentration (Cmax), qualifying uHeart as a fit-to-purpose preclinical tool for cardiotoxicity studies.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Células-Tronco Pluripotentes Induzidas , Dispositivos Lab-On-A-Chip , Síndrome do QT Longo , Humanos , Cardiotoxicidade , Avaliação Pré-Clínica de Medicamentos/métodos , Canais Iônicos , Síndrome do QT Longo/induzido quimicamente , Miócitos Cardíacos , Preparações Farmacêuticas
2.
Biomed Mater ; 16(4)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34030149

RESUMO

A microfluidic technique is presented for micropatterning protein domains and cell cultures within permanently bonded organs-on-chip devices. This method is based on the use of polydimethylsiloxane layers coupled with the plasma ablation technique for selective protein removal. We show how this technique can be employed to generate a multi-organin vitromodel directly within a microscale platform suitable for pharmacokinetic-based drug screening. We miniaturized a liver model based on micropatterned co-cultures in dual-compartment microfluidic devices. The cytotoxic effect of liver-metabolized Tegafur on colon cancer cell line was assessed using two microfluidic devices where microgrooves and valves systems are used to model drug diffusion between culture compartments. The platforms can reproduce the metabolism of Tegafur in the liver, thus killing colon cancer cells. The proposed plasma-enhanced microfluidic protein patterning method thus successfully combines the ability to generate precise cell micropatterning with the intrinsic advantages of microfluidics in cell biology.


Assuntos
Dispositivos Lab-On-A-Chip , Neoplasias Hepáticas/metabolismo , Modelos Biológicos , Análise Serial de Tecidos/métodos , Biotecnologia , Sobrevivência Celular , Dimetilpolisiloxanos , Avaliação Pré-Clínica de Medicamentos , Desenho de Equipamento , Humanos , Técnicas Analíticas Microfluídicas
3.
Biosensors (Basel) ; 10(9)2020 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-32872228

RESUMO

Organs-on-chip (OoC), often referred to as microphysiological systems (MPS), are advanced in vitro tools able to replicate essential functions of human organs. Owing to their unprecedented ability to recapitulate key features of the native cellular environments, they represent promising tools for tissue engineering and drug screening applications. The achievement of proper functionalities within OoC is crucial; to this purpose, several parameters (e.g., chemical, physical) need to be assessed. Currently, most approaches rely on off-chip analysis and imaging techniques. However, the urgent demand for continuous, noninvasive, and real-time monitoring of tissue constructs requires the direct integration of biosensors. In this review, we focus on recent strategies to miniaturize and embed biosensing systems into organs-on-chip platforms. Biosensors for monitoring biological models with metabolic activities, models with tissue barrier functions, as well as models with electromechanical properties will be described and critically evaluated. In addition, multisensor integration within multiorgan platforms will be further reviewed and discussed.


Assuntos
Técnicas Biossensoriais , Dispositivos Lab-On-A-Chip , Monitorização Fisiológica , Avaliação Pré-Clínica de Medicamentos , Humanos , Procedimentos Analíticos em Microchip , Microfluídica , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Engenharia Tecidual
4.
Neuroscience ; 416: 88-99, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31400485

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting the corticospinal tract and leading to motor neuron death. According to a recent study, magnetic resonance imaging-visible changes suggestive of neurodegeneration seem absent in the motor cortex of G93A-SOD1 ALS mice. However, it has not yet been ascertained whether the cortical neural activity is intact, or alterations are present, perhaps even from an early stage. Here, cortical neurons from this model were isolated at post-natal day 1 and cultured on multielectrode arrays. Their activity was studied with a comprehensive pool of neurophysiological analyses probing excitability, criticality and network architecture, alongside immunocytochemistry and molecular investigations. Significant hyperexcitability was visible through increased network firing rate and bursting, whereas topological changes in the synchronization patterns were apparently absent. The number of dendritic spines was increased, accompanied by elevated transcriptional levels of the DLG4 gene, NMDA receptor 1 and the early pro-apoptotic APAF1 gene. The extracellular Na+, Ca2+, K+ and Cl- concentrations were elevated, pointing to perturbations in the culture micro-environment. Our findings highlight remarkable early changes in ALS cortical neuron activity and physiology. These changes suggest that the causative factors of hyperexcitability and associated toxicity could become established much earlier than the appearance of disease symptoms, with implications for the discovery of new hypothetical therapeutic targets.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Córtex Motor/patologia , Neurônios Motores/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Morte Celular/fisiologia , Modelos Animais de Doenças , Camundongos Transgênicos , Doenças Neurodegenerativas/patologia , Superóxido Dismutase/metabolismo
5.
Med Eng Phys ; 48: 31-38, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28869117

RESUMO

Thrombus formation is a major adverse event affecting patients implanted with ventricular assist devices (VADs). Despite anti-thrombotic drug administration, thrombotic events remain frequent within the first year post-implantation. Platelet activation (PA) is an essential process underling thrombotic adverse events in VAD systems. Indeed, abnormal shear forces, correlating with specific flow trajectories of VADs, are strong agonists mediating PA. To date, the ability to determine efficacy of anti-platelet (AP) agents under shear stress conditions is limited. Here, we present a novel microfluidic platform designed to replicate shear stress patterns of a clinical VAD, and use it to compare the efficacy of two AP agents in vitro. Gel-filtered platelets were incubated with i) acetylsalicylic acid (ASA) and ii) ticagrelor, at two different concentrations (ASA: 125 and 250 µM; ticagrelor: 250 and 500 nM) and were circulated in the VAD-emulating microfluidic platform using a peristaltic pump. GFP was collected after 4 and 52 repetitions of exposure to the VAD shear pattern and tested for shear-mediated PA. ASA significantly inhibited PA only at 2-fold higher concentration (250 µM) than therapeutic dose (125 µM). The effect of ticagrelor was not dependent on drug concentration, and did not show significant inhibition with respect to untreated control. This study demonstrates the potential use of microfluidic platforms as means of testing platelet responsiveness and AP drug efficacy under complex and realistic VAD-like shear stress conditions.


Assuntos
Avaliação Pré-Clínica de Medicamentos/instrumentação , Coração Auxiliar , Dispositivos Lab-On-A-Chip , Inibidores da Agregação Plaquetária/farmacologia , Resistência ao Cisalhamento , Adenosina/análogos & derivados , Adenosina/farmacologia , Aspirina/farmacologia , Sonicação , Ticagrelor
6.
Methods Mol Biol ; 1612: 303-323, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28634953

RESUMO

The design of innovative tools for generating physiologically relevant three-dimensional (3D) in vitro models has been recently recognized as a fundamental step to study cell responses and long-term tissue functionalities thanks to its ability to recapitulate the complexity and the dimensional scale of the cellular microenvironment, while directly integrating high-throughput and automatic screening capabilities.This chapter addresses the development of a poly(dimethylsiloxane)-based microfluidic platform to (1) generate and culture 3D cellular microaggregates under continuous flow perfusion while (2) conditioning them with different combinations/concentrations of soluble factors (i.e., growth factors, morphogens or drug molecules), in a high-throughput fashion. The proposed microfluidic system thus represents a promising tool for establishing innovative high-throughput models for drug screening, investigation of tissues morphogenesis, and optimization of tissue engineering protocols.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Técnicas Analíticas Microfluídicas/instrumentação , Animais , Técnicas de Cultura de Células/instrumentação , Dimetilpolisiloxanos/química , Avaliação Pré-Clínica de Medicamentos , Humanos , Engenharia Tecidual
7.
Biomaterials ; 110: 45-59, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27710832

RESUMO

Engineering cardiac tissues and organ models remains a great challenge due to the hierarchical structure of the native myocardium. The need of integrating blood vessels brings additional complexity, limiting the available approaches that are suitable to produce integrated cardiovascular organoids. In this work we propose a novel hybrid strategy based on 3D bioprinting, to fabricate endothelialized myocardium. Enabled by the use of our composite bioink, endothelial cells directly bioprinted within microfibrous hydrogel scaffolds gradually migrated towards the peripheries of the microfibers to form a layer of confluent endothelium. Together with controlled anisotropy, this 3D endothelial bed was then seeded with cardiomyocytes to generate aligned myocardium capable of spontaneous and synchronous contraction. We further embedded the organoids into a specially designed microfluidic perfusion bioreactor to complete the endothelialized-myocardium-on-a-chip platform for cardiovascular toxicity evaluation. Finally, we demonstrated that such a technique could be translated to human cardiomyocytes derived from induced pluripotent stem cells to construct endothelialized human myocardium. We believe that our method for generation of endothelialized organoids fabricated through an innovative 3D bioprinting technology may find widespread applications in regenerative medicine, drug screening, and potentially disease modeling.


Assuntos
Bioimpressão/métodos , Células Endoteliais , Miocárdio , Organoides/crescimento & desenvolvimento , Impressão Tridimensional , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/química , Células Endoteliais/citologia , Humanos , Hidrogéis/química , Microfibrilas/química , Miócitos Cardíacos/química , Miócitos Cardíacos/metabolismo , Organoides/química , Organoides/metabolismo , Medicina Regenerativa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA