Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biotechnol Bioeng ; 118(8): 3128-3137, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34019719

RESUMEN

Heart-on-chip is an unprecedented technology for recapitulating key biochemical and biophysical cues in cardiac pathophysiology. Several designs have been proposed to improve its ability to mimic the native tissue and establish it as a reliable research platform. However, despite mimicking one of most vascularized organs, reliable strategies to deliver oxygen and substrates to densely packed constructs of metabolically demanding cells remain unsettled. Herein, we describe a new heart-on-chip platform with precise fluid control, integrating an on-chip peristaltic pump, allowing automated and fine control over flow on channels flanking a 3D cardiac culture. The application of distinct flow rates impacted on temporal dynamics of microtissue structural and transcriptional maturation, improving functional performance. Moreover, a widespread transcriptional response was observed, suggesting flow-mediated activation of critical pathways of cardiomyocyte structural and functional maturation and inhibition of cardiomyocyte hypoxic injury. In conclusion, the present design represents an important advance in bringing engineered cardiac microtissues closer to the native heart, overcoming traditional bulky off-chip fluid handling systems, improving microtissue performance, and matching oxygen and energy substrate requirements of metabolically active constructs, avoiding cellular hypoxia. Distinct flow patterns differently impact on microtissue performance and gene expression program.


Asunto(s)
Bombas de Infusión , Dispositivos Laboratorio en un Chip , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Perfusión , Animales , Hipoxia de la Célula , Ratas , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos
2.
Biotechnol Bioeng ; 115(3): 775-784, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29197187

RESUMEN

The grail of gene delivery is the development of delivery vectors as effective and non-cytotoxic as possible. In this regard, there is an urgent need of new tools for the straightforward and quantitative assessment of transfection efficiency and cytotoxicity simultaneously. We herein reported the development and validation of an easy-to-use lab-on-chip platform to perform cell transfection assays for unbiased, high-throughput selection of more and more effective gene delivery vectors by using two commercially sourced lipids, Lipofectamine 2000® and FuGene® 6. A single PDMS-layer platform was endowed with: i) a chaotic serial dilution generator, designed for the automatic generation of a linear lipoplex dilution (from 100% to 0% with 25% steps) independently delivered to; and ii) the downstream culture and transfection module consisting in five units, each composed of 33 serially connected and fluidically connected culture chambers for trapping small populations of ≈10 cells/chamber. In the absence of any transfectant, cells spread and duplicated up to 2 days. Besides, cells were transfected with EGFP-encoding reporter gene. The very facile visual inspection of the microdevice by means of a microscope and a semi-automated analytical method allowed pinpointing the best transfection conditions in terms of efficiency, cytotoxicity, cell doubling rates, and morphological changes at once.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Dispositivos Laboratorio en un Chip , Vectores Genéticos/genética , Vectores Genéticos/farmacología , Células HeLa , Humanos
3.
Molecules ; 21(9)2016 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-27571058

RESUMEN

In the last few years microfluidics and microfabrication technique principles have been extensively exploited for biomedical applications. In this framework, organs-on-a-chip represent promising tools to reproduce key features of functional tissue units within microscale culture chambers. These systems offer the possibility to investigate the effects of biochemical, mechanical, and electrical stimulations, which are usually applied to enhance the functionality of the engineered tissues. Since the functionality of muscle tissues relies on the 3D organization and on the perfect coupling between electrochemical stimulation and mechanical contraction, great efforts have been devoted to generate biomimetic skeletal and cardiac systems to allow high-throughput pathophysiological studies and drug screening. This review critically analyzes microfluidic platforms that were designed for skeletal and cardiac muscle tissue engineering. Our aim is to highlight which specific features of the engineered systems promoted a typical reorganization of the engineered construct and to discuss how promising design solutions exploited for skeletal muscle models could be applied to improve cardiac tissue models and vice versa.


Asunto(s)
Dispositivos Laboratorio en un Chip , Modelos Biológicos , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Ingeniería de Tejidos/métodos , Animales , Humanos , Músculo Esquelético/citología , Miocardio/citología
4.
Adv Healthc Mater ; 13(4): e2301481, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37941521

RESUMEN

Cardiac fibrosis is one of the main causes of heart failure, significantly contributing to mortality. The discovery and development of effective therapies able to heal fibrotic pathological symptoms thus remain of paramount importance. Micro-physiological systems (MPS) are recently introduced as promising platforms able to accelerate this finding. Here a 3D in vitro model of human cardiac fibrosis, named uScar, is developed by imposing a cyclic mechanical stimulation to human atrial cardiac fibroblasts (AHCFs) cultured in a 3D beating heart-on-chip and exploited to screen drugs and advanced therapeutics. The sole provision of a cyclic 10% uniaxial strain at 1 Hz to the microtissues is sufficient to trigger fibrotic traits, inducing a consistent fibroblast-to-myofibroblast transition and an enhanced expression and production of extracellular matrix (ECM) proteins. Standard of care anti-fibrotic drugs (i.e., Pirfenidone and Tranilast) are confirmed to be efficient in preventing the onset of fibrotic traits in uScar. Conversely, the mechanical stimulation applied to the microtissues limit the ability of a miRNA therapy to directly reprogram fibroblasts into cardiomyocytes (CMs), despite its proved efficacy in 2D models. Such results demonstrate the importance of incorporating in vivo-like stimulations to generate more representative 3D in vitro models able to predict the efficacy of therapies in patients.


Asunto(s)
Cardiomiopatías , Miocitos Cardíacos , Humanos , Miocitos Cardíacos/metabolismo , Cardiomiopatías/metabolismo , Fibrosis , Fibroblastos/metabolismo , Miofibroblastos/patología , Proteínas de la Matriz Extracelular/metabolismo , Miocardio/metabolismo
5.
Toxicol Sci ; 191(1): 47-60, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36226800

RESUMEN

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.


Asunto(s)
Evaluación Preclínica de Medicamentos , Células Madre Pluripotentes Inducidas , Dispositivos Laboratorio en un Chip , Síndrome de QT Prolongado , Humanos , Cardiotoxicidad , Evaluación Preclínica de Medicamentos/métodos , Canales Iónicos , Síndrome de QT Prolongado/inducido químicamente , Miocitos Cardíacos , Preparaciones Farmacéuticas
6.
Methods Mol Biol ; 2373: 133-157, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34520011

RESUMEN

Modeling human cardiac tissues in vitro is essential to elucidate the biological mechanisms related to the heart physiopathology, possibly paving the way for new treatments. Organs-on-chips have emerged as innovative tools able to recreate tissue-specific microenvironments, guiding the development of miniaturized models and offering the opportunity to directly analyze functional readouts. Here we describe the fabrication and operational procedures for the development of a heart-on-chip model, reproducing cardiac biomimetic microenvironment. The device provides 3D cardiac microtissue with a synchronized electromechanical stimulation to support the tissue development. We additionally describe procedures for characterizing tissue evolution and functionality through immunofluorescence, real time qPCR, calcium imaging and microtissue contractility investigations.


Asunto(s)
Corazón , Biomimética , Calcio , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
7.
Biophys Rev ; 14(3): 735, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35791385

RESUMEN

[This corrects the article DOI: 10.1007/s12551-021-00841-6.].

8.
Lab Chip ; 23(1): 136-145, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36477137

RESUMEN

Transfection describes the delivery of exogenous nucleic acids (NAs) to cells utilizing non-viral means. In the last few decades, scientists have been doing their utmost to design ever more effective transfection reagents. These are eventually mixed with NAs to give rise to gene delivery complexes, which must undergo characterization, testing, and further refinement through the sequential reiteration of these steps. Unfortunately, although microfluidics offers distinct advantages over the canonical approaches to preparing particles, the systems available do not address the most frequent and practical quest for the simultaneous generation of multiple polymer-to-NA ratios (N/Ps). Herein, we developed a user-friendly microfluidic cartridge to repeatably prepare non-viral gene delivery particles and screen across a range of seven N/Ps at once or significant volumes of polyplexes at a given N/P. The microchip is equipped with a chaotic serial dilution generator for the automatic linear dilution of the polymer to the downstream area, which encompasses the NA divider to dispense equal amounts of DNA to the mixing area, enabling the formation of particles at seven N/Ps eventually collected in individual built-in tanks. This is the first example of a stand-alone microfluidic cartridge for the fast and repeatable preparation of non-viral gene delivery complexes at different N/Ps and their storage.


Asunto(s)
Técnicas de Transferencia de Gen , Microfluídica , Transfección , ADN , Polímeros
9.
Biomaterials ; 280: 121248, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34794827

RESUMEN

Hemodynamics play a central role in the health and disease of the coronary and peripheral vascular systems. Vessel-lining endothelial cells are known mechanosensors, responding to disturbances in flow - with mechanosensitivity hypothesized to change in response to metabolic demands. The health of our smallest microvessels have been lauded as a prognostic marker for cardiovascular health. Yet, despite numerous animal models, studying these small vessels has proved difficult. Microfluidic technologies have allowed a number of 3D vascular models to be developed and used to investigate human vessels. Here, two such systems are employed for examining 1) interstitial flow effects on neo-vessel formation, and 2) the effects of flow-conditioning on vascular remodeling following sustained static culture. Interstitial flow is shown to enhance early vessel formation via significant remodeling of vessels and interconnected tight junctions of the endothelium. In formed vessels, continuous flow maintains a stable vascular diameter and causes significant remodeling, contrasting the continued anti-angiogenic decline of statically cultured vessels. This study is the first to couple complex 3D computational flow distributions and microvessel remodeling from microvessels grown on-chip (exposed to flow or no-flow conditions). Flow-conditioned vessels (WSS < 1Pa for 30 µm vessels) increase endothelial barrier function, result in significant changes in gene expression and reduce reactive oxygen species and anti-angiogenic cytokines. Taken together, these results demonstrate microvessel mechanosensitivity to flow-conditioning, which limits deleterious vessel regression in vitro, and could have implications for future modeling of reperfusion/no-flow conditions.


Asunto(s)
Capilares , Células Endoteliales , Animales , Hemodinámica , Humanos , Microfluídica , Microvasos
10.
Front Physiol ; 12: 735915, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34690810

RESUMEN

Fibrosis, a hallmark of many cardiac and pulmonary diseases, is characterized by excess deposition of extracellular matrix proteins and increased tissue stiffness. This serious pathologic condition is thought to stem majorly from local stromal cell activation. Most studies have focused on the role of fibroblasts; however, the endothelium has been implicated in fibrosis through direct and indirect contributions. Here, we present a 3D vascular model to investigate vessel-stroma crosstalk in normal conditions and following induced fibrosis. Human-induced pluripotent stem cell-derived endothelial cells (hiPSC-ECs) are co-cultured with (and without) primary human cardiac and lung fibroblasts (LFs) in a microfluidic device to generate perfusable microvasculature in cardiac- and pulmonary-like microenvironments. Endothelial barrier function, vascular morphology, and matrix properties (stiffness and diffusivity) are differentially impacted by the presence of stromal cells. These vessels (with and without stromal cells) express inflammatory cytokines, which could induce a wound-healing state. Further treatment with transforming growth factor-ß (TGF-ß) induced varied fibrotic phenotypes on-chip, with LFs resulting in increased stiffness, lower MMP activity, and increased smooth muscle actin expression. Taken together, our work demonstrates the strong impact of stromal-endothelial interactions on vessel formation and extravascular matrix regulation. The role of TGF-ß is shown to affect co-cultured microvessels differentially and has a severe negative impact on the endothelium without stromal cell support. Our human 3D in vitro model has the potential to examine anti-fibrotic therapies on patient-specific hiPSCs in the future.

11.
Biophys Rev ; 13(5): 717-727, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34765047

RESUMEN

The most advanced in vitro cardiac models are today based on the use of induced pluripotent stem cells (iPSCs); however, the maturation of cardiomyocytes (CMs) has not yet been fully achieved. Therefore, there is a rising need to move towards models capable of promoting an adult-like cardiomyocytes phenotype. Many strategies have been applied such as co-culture of cardiomyocytes, with fibroblasts and endothelial cells, or conditioning them through biochemical factors and physical stimulations. Here, we focus on mechanical stimulation as it aims to mimic the different mechanical forces that heart receives during its development and the post-natal period. We describe the current strategies and the mechanical properties necessary to promote a positive response in cardiac tissues from different cell sources, distinguishing between passive stimulation, which includes stiffness, topography and static stress and active stimulation, encompassing cyclic strain, compression or perfusion. We also highlight how mechanical stimulation is applied in disease modelling.

12.
Lab Chip ; 21(21): 4177-4195, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34545378

RESUMEN

Cardiac fibrosis is a maladaptive remodeling of the myocardium hallmarked by contraction impairment and excessive extracellular matrix deposition (ECM). The disease progression, nevertheless, remains poorly understood and present treatments are not capable of controlling the scarring process. This is partly due to the absence of physiologically relevant, easily operable, and low-cost in vitro models, which are of the utmost importance to uncover pathological mechanisms and highlight possible targets for anti-fibrotic therapies. In classic models, fibrotic features are usually obtained using substrates with scar mimicking stiffness and/or supplementation of morphogens such as transforming growth factor ß1 (TGF-ß1). Qualities such as the interplay between activated fibroblasts (FBs) and cardiomyocytes (CMs), or the mechanically active, three-dimensional (3D) environment, are, however, neglected or obtained at the expense of the number of experimental replicates achievable. To overcome these shortcomings, we engineered a micro-physiological system (MPS) where multiple 3D cardiac micro-tissues can be subjected to cyclical stretching simultaneously. Up to six different biologically independent samples are incorporated in a single device, increasing the experimental throughput and paving the way for higher yielding drug screening campaigns. The newly developed MPS was used to co-culture different ratios of neonatal rat CMs and FBs, investigating the role of CMs in the modulation of fibrosis traits, without the addition of morphogens, and in soft substrates. The expression of contractile stress fibers and of degradative enzymes, as well as the deposition of fibronectin and type I collagen were superior in microtissues with a low amount of CMs. Moreover, high CM-based microconstructs simulating a ratio similar to that of healthy tissues, even if subjected to both cyclic stretch and TGF-ß1, did not show any of the investigated fibrotic signs, indicating a CM fibrosis modulating effect. Overall, this in vitro fibrosis model could help to uncover new pathological aspects studying, with mid-throughput and in a mechanically active, physiologically relevant environment, the crosstalk between the most abundant cell types involved in fibrosis.


Asunto(s)
Fibroblastos , Miocitos Cardíacos , Animales , Células Cultivadas , Matriz Extracelular , Fibroblastos/patología , Fibrosis , Ratas , Factor de Crecimiento Transformador beta1
13.
Biofabrication ; 13(4)2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34139683

RESUMEN

The synovium of osteoarthritis (OA) patients can be characterized by an abnormal accumulation of macrophages originating from extravasated monocytes. Since targeting monocyte extravasation may represent a promising therapeutic strategy, our aim was to develop an organotypic microfluidic model recapitulating this process. Synovium and cartilage were modeled by hydrogel-embedded OA synovial fibroblasts and articular chondrocytes separated by a synovial fluid channel. The synovium compartment included a perfusable endothelialized channel dedicated to monocyte injection. Monocyte extravasation in response to chemokines and OA synovial fluid was quantified. The efficacy of chemokine receptor antagonists, RS-504393 (CCR2 antagonist) and Cenicriviroc (CCR2/CCR5 antagonist) in inhibiting extravasation was tested pre-incubating monocytes with the antagonists before injection. After designing and fabricating the chip, culture conditions were optimized to achieve an organotypic model including synovial fibroblasts, articular chondrocytes, and a continuous endothelial monolayer expressing intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. A significantly higher number of monocytes extravasated in response to the chemokine mix (p< 0.01) and OA synovial fluid (p< 0.01), compared to a control condition. In both cases, endothelium pre-activation enhanced monocyte extravasation. The simultaneous blocking of CCR2 and CCR5 proved to be more effective (p< 0.001) in inhibiting monocyte extravasation in response to OA synovial fluid than blocking of CCR2 only (p< 0.01). The study of extravasation in the model provided direct evidence that OA synovial fluid induces monocytes to cross the endothelium and invade the synovial compartment. The model can be exploited either to test molecules antagonizing this process or to investigate the effect of extravasated monocytes on synovium and cartilage cells.


Asunto(s)
Monocitos , Membrana Sinovial , Cartílago Articular , Humanos , Microfluídica , Osteoartritis , Líquido Sinovial
14.
Biofabrication ; 13(3)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33561845

RESUMEN

Cardiac toxicity still represents a common adverse outcome causing drug attrition and post-marketing withdrawal. The development of relevantin vitromodels resembling the human heart recently opened the path towards a more accurate detection of drug-induced human cardiac toxicity early in the drug development process. Organs-on-chip have been proposed as promising tools to recapitulatein vitrothe key aspects of thein vivocardiac physiology and to provide a means to directly analyze functional readouts. In this scenario, a new device capable of continuous monitoring of electrophysiological signals from functionalin vitrohuman hearts-on-chip is here presented. The development of cardiac microtissues was achieved through a recently published method to control the mechanical environment, while the introduction of a technology consisting in micro-electrode coaxial guides allowed to conduct direct and non-destructive electrophysiology studies. The generated human cardiac microtissues exhibited synchronous spontaneous beating, as demonstrated by multi-point and continuous acquisition of cardiac field potential, and expression of relevant genes encoding for cardiac ion-channels. A proof-of-concept pharmacological validation on three drugs proved the proposed model to potentially be a powerful tool to evaluate functional cardiac toxicity.


Asunto(s)
Fenómenos Electrofisiológicos , Corazón , Electricidad , Electrodos , Corazón/fisiología , Humanos , Miocitos Cardíacos
15.
Sci Rep ; 8(1): 16944, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30446711

RESUMEN

Tissue engineering strategies have been extensively exploited to generate functional cardiac patches. To maintain cardiac functionality in vitro, bioreactors have been designed to provide perfusion and electrical stimulation, alone or combined. However, due to several design limitations the integration of optical systems to assess cardiac maturation level is still missing within these platforms. Here we present a bioreactor culture chamber that provides 3D cardiac constructs with a bidirectional interstitial perfusion and biomimetic electrical stimulation, allowing direct cellular optical monitoring and contractility test. The chamber design was optimized through finite element models to house an innovative scaffold anchoring system to hold and to release it for the evaluation of tissue maturation and functionality by contractility tests. Neonatal rat cardiac fibroblasts subjected to a combined perfusion and electrical stimulation showed positive cell viability over time. Neonatal rat cardiomyocytes were successfully monitored for the entire culture period to assess their functionality. The combination of perfusion and electrical stimulation enhanced patch maturation, as evidenced by the higher contractility, the enhanced beating properties and the increased level of cardiac protein expression. This new multifunctional bioreactor provides a relevant biomimetic environment allowing for independently culturing, real-time monitoring and testing up to 18 separated patches.


Asunto(s)
Reactores Biológicos , Fibroblastos/citología , Miocitos Cardíacos/citología , Ingeniería de Tejidos/métodos , Animales , Animales Recién Nacidos , Células Cultivadas , Estimulación Eléctrica , Fibroblastos/metabolismo , Expresión Génica , Corazón/fisiología , Contracción Miocárdica/fisiología , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Perfusión/métodos , Ratas , Ingeniería de Tejidos/instrumentación
16.
Methods Cell Biol ; 146: 69-84, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30037467

RESUMEN

With the increasing attention on cardiovascular disorders and the current inability of pre-clinical models to accurately predict human physiology, the need for advanced and reliable heart in vitro models is paramount. Microfabrication technologies provide potential solutions in the organs-on-chip systems: microengineered devices where cell cultures can be hosted and cultured to develop three-dimensional models or microtissues with high similarity to human physiology. We here described the fabrication and operation procedures for a beating heart-on-a-chip. The device features a culture region for a 3D cardiac microtissue and a system for applying tuned mechanical stimulation during culture to improve cardiac development. We additionally describe procedures for characterizing tissue maturation via immunofluorescence and functional evaluations of microtissue contractility.


Asunto(s)
Corazón/fisiología , Dispositivos Laboratorio en un Chip , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos , Humanos , Microfluídica
17.
APL Bioeng ; 2(4): 046102, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31069324

RESUMEN

Organs-on-chip technology has recently emerged as a promising tool to generate advanced cardiac tissue in vitro models, by recapitulating key physiological cues of the native myocardium. Biochemical, mechanical, and electrical stimuli have been investigated and demonstrated to enhance the maturation of cardiac constructs. However, the combined application of such stimulations on 3D organized constructs within a microfluidic platform was not yet achieved. For this purpose, we developed an innovative microbioreactor designed to provide a uniform electric field and cyclic uniaxial strains to 3D cardiac microtissues, recapitulating the complex electro-mechanical environment of the heart. The platform encompasses a compartment to confine and culture cell-laden hydrogels, a pressure-actuated chamber to apply a cyclic uniaxial stretch to microtissues, and stainless-steel electrodes to accurately regulate the electric field. The platform was exploited to investigate the effect of two different electrical stimulation patterns on cardiac microtissues from neonatal rat cardiomyocytes: a controlled electric field [5 V/cm, or low voltage (LV)] and a controlled current density [74.4 mA/cm2, or high voltage (HV)]. Our results demonstrated that LV stimulation enhanced the beating properties of the microtissues. By fully exploiting the platform, we combined the LV electrical stimulation with a physiologic mechanical stretch (10% strain) to recapitulate the key cues of the native cardiac microenvironment. The proposed microbioreactor represents an innovative tool to culture improved miniaturized cardiac tissue models for basic research studies on heart physiopathology and for drug screening.

18.
Integr Biol (Camb) ; 10(3): 174-183, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29532839

RESUMEN

In vitro cardiac models able to mimic the fibrotic process are paramount to develop an effective anti-fibrosis therapy that can regulate fibroblast behaviour upon myocardial injury. In previously developed in vitro models, typical fibrosis features were induced by using scar-like stiffness substrates and/or potent morphogen supplementation in monolayer cultures. In our model, we aimed to mimic in vitro a fibrosis-like environment by applying cyclic stretching of cardiac fibroblasts embedded in three-dimensional fibrin-hydrogels alone. Using a microfluidic device capable of delivering controlled cyclic mechanical stretching (10% strain at 1 Hz), some of the main fibrosis hallmarks were successfully reproduced in 7 days. Cyclic strain indeed increased cell proliferation, extracellular matrix (ECM) deposition (e.g. type-I-collagen, fibronectin) and its stiffness, forming a scar-like tissue with superior quality compared to the supplementation of TGFß1 alone. Taken together, the observed findings resemble some of the key steps in the formation of a scar: (i) early fibroblast proliferation, (ii) later phenotype switch into myofibroblasts, (iii) ECM deposition and (iv) stiffening. This in vitro scar-on-a-chip model represents a big step forward to investigate the early mechanisms possibly leading later to fibrosis without any possible confounding supplementation of exogenous potent morphogens.


Asunto(s)
Cicatriz/patología , Fibroblastos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Animales , Animales Recién Nacidos , Proliferación Celular , Colágeno Tipo I/metabolismo , Dimetilpolisiloxanos/química , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Fibrosis/patología , Humanos , Hidrogeles , Técnicas In Vitro , Dispositivos Laboratorio en un Chip , Microfluídica , Infarto del Miocardio/patología , Miofibroblastos/metabolismo , Fenotipo , Ratas , Estrés Mecánico , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas
19.
Methods Mol Biol ; 1612: 303-323, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28634953

RESUMEN

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.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Técnicas Analíticas Microfluídicas/instrumentación , Animales , Técnicas de Cultivo de Célula/instrumentación , Dimetilpolisiloxanos/química , Evaluación Preclínica de Medicamentos , Humanos , Ingeniería de Tejidos
20.
Adv Healthc Mater ; 6(10)2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28267277

RESUMEN

A novel technique is presented for molding and culturing composite 3D cellular constructs within microfluidic channels. The method is based on the use of removable molding polydimethylsiloxane (PDMS) inserts, which allow to selectively and incrementally generate composite 3D constructs featuring different cell types and/or biomaterials, with a high spatial control. The authors generate constructs made of either stacked hydrogels, with uniform horizontal interfaces, or flanked hydrogels with vertical interfaces. The authors also show how this technique can be employed to create custom-shaped endothelial barriers and monolayers directly interfaced with 3D cellular constructs. This method dramatically improves the significance of in vitro 3D biological models, enhancing mimicry and enabling for controlled studies of complex biological districts.


Asunto(s)
Microfluídica/métodos , Materiales Biocompatibles/química , Técnicas de Cultivo de Célula , Dimetilpolisiloxanos/química , Hidrogeles/química , Dispositivos Laboratorio en un Chip
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA