Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Adv Healthc Mater ; 13(7): e2302454, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38253407

RESUMO

The multifactorial nature of inflammatory bowel disease (IBD) necessitates reliable and practical experimental models to elucidate its etiology and pathogenesis. To model the intestinal microenvironment at the onset of IBD in vitro, it is important to incorporate relevant cellular and noncellular components before inducing stepwise pathogenic developments. A novel intestine-on-chip system for investigating multiple aspects of IBD's immunopathogenesis is presented. The system includes an array of tight and polarized barrier models formed from intestinal epithelial cells on an in-vivo-like subepithelial matrix within one week. The dynamic remodeling of the subepithelial matrix by cells or their secretome demonstrates the physiological relevance of the on-chip barrier models. The system design enables introduction of various immune cell types and inflammatory stimuli at specific locations in the same barrier model, which facilitates investigations of the distinct roles of each cell type in intestinal inflammation development. It is showed that inflammatory behavior manifests in an upregulated expression of inflammatory markers and cytokines (TNF-α). The neutralizing effect of the anti-inflammatory antibody Infliximab on levels of TNF-α and its inducible cytokines could be explicitly shown. Overall, an innovative approach to systematically developing a microphysiological system to comprehend immune-system-mediated disorders of IBD and to identify new therapeutic strategies is presented.


Assuntos
Doenças Inflamatórias Intestinais , Fator de Necrose Tumoral alfa , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Mucosa Intestinal/metabolismo , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/patologia , Intestinos/patologia , Citocinas/metabolismo
2.
Lab Chip ; 23(22): 4834-4847, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37853793

RESUMO

Integrating flowing cells, such as immune cells or circulating tumour cells, within a microphysiological system is crucial for body-on-a-chip applications. However, ensuring unimpeded recirculation of cells is a significant challenge. Closed microfluidic devices have a no-slip boundary condition along channel walls and a defined chip geometry (laminar flow) that hinders the ability to freely control cell flow. Open microfluidic devices, where the bottom device boundary is an air-liquid interface (ALI), e.g., hanging drop networks (HDNs), offer the advantage of an easily-actuatable fluid-phase geometry, where cells can either flow or stagnate. In this paper, we optimized a hanging-drop-integrated pneumatic-pump system for closed-loop recirculation of particles (i.e., beads or cells). Experiments with both beads and cells in cell culture medium initially resulted in particle stagnation, which was suggestive of a pseudo-no-slip boundary condition at the ALI. Transmission electron microscopy and dynamic light scattering measurements of the ALI suggested that aggregation of submicron-scale cell-culture-medium components is the cause of the pseudo-no-slip boundary condition. We used the finite element method to study the forces on particles at the ALI and to optimize HDN design (drop aperture) and operation (drop height) parameters. Based on this analysis, we report a phase diagram delineating the conditions for free flow or stagnation of particles at the ALI of hanging drops. Using our experimental setup with 3.5 mm drop apertures, we conducted particle flow experiments while actuating drop heights. We confirmed the ability to control the flow or stagnation of particles by actuating the height of hanging drops: a drop height over 300 µm led to particle stagnation and a drop height under 300 µm allowed for particle flow. This particle-flow control, combined with the ease of integrating scaffold-free organ models (microtissues or organoids) in HDNs, constitutes the basis for an experimental setup enabling the control of the residence time of single cells around 3D organ models.


Assuntos
Técnicas de Cultura de Células , Esferoides Celulares , Técnicas de Cultura de Células/métodos , Movimento Celular , Dispositivos Lab-On-A-Chip , Sistemas Microfisiológicos
3.
Adv Healthc Mater ; 12(6): e2202506, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36651229

RESUMO

Despite increasing survival rates of pediatric leukemia patients over the past decades, the outcome of some leukemia subtypes has remained dismal. Drug sensitivity and resistance testing on patient-derived leukemia samples provide important information to tailor treatments for high-risk patients. However, currently used well-based drug screening platforms have limitations in predicting the effects of prodrugs, a class of therapeutics that require metabolic activation to become effective. To address this issue, a microphysiological drug-testing platform is developed that enables co-culturing of patient-derived leukemia cells, human bone marrow mesenchymal stromal cells, and human liver microtissues within the same microfluidic platform. This platform also enables to control the physical interaction between the diverse cell types. Herein, it is made possible to recapitulate hepatic prodrug activation of ifosfamide in their platform, which is very difficult in traditional well-based assays. By testing the susceptibility of primary patient-derived leukemia samples to the prodrug ifosfamide, sample-specific sensitivities to ifosfamide in primary leukemia samples are identified. The microfluidic platform is found to enable the recapitulation of physiologically relevant conditions and the testing of prodrugs including short-lived and unstable metabolites. The platform holds great potential for clinical translation and precision chemotherapy selection.


Assuntos
Leucemia , Pró-Fármacos , Humanos , Criança , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Pró-Fármacos/metabolismo , Ifosfamida/farmacologia , Ifosfamida/uso terapêutico , Ifosfamida/metabolismo , Leucemia/metabolismo , Técnicas de Cocultura , Fígado/metabolismo
4.
Micromachines (Basel) ; 13(7)2022 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-35888941

RESUMO

Microfluidic-drop networks consist of several stable drops-interconnected through microfluidic channels-in which organ models can be cultured long-term. Drop networks feature a versatile configuration and an air-liquid interface (ALI). This ALI provides ample oxygenation, rapid liquid turnover, passive degassing, and liquid-phase stability through capillary pressure. Mathematical modeling, e.g., by using computational fluid dynamics (CFD), is a powerful tool to design drop-based microfluidic devices and to optimize their operation. Although CFD is the most rigorous technique to model flow, it falls short in terms of computational efficiency. Alternatively, the hydraulic-electric analogy is an efficient "first-pass" method to explore the design and operation parameter space of microfluidic-drop networks. However, there are no direct electric analogs to a drop, due to the nonlinear nature of the capillary pressure of the ALI. Here, we present a circuit-based model of hanging- and standing-drop compartments. We show a phase diagram describing the nonlinearity of the capillary pressure of a hanging drop. This diagram explains how to experimentally ensure drop stability. We present a methodology to find flow rates and pressures within drop networks. Finally, we review several applications, where the method, outlined in this paper, was instrumental in optimizing design and operation.

5.
Front Immunol ; 12: 781337, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925361

RESUMO

Existing first-line cancer therapies often fail to cope with the heterogeneity and complexity of cancers, so that new therapeutic approaches are urgently needed. Among novel alternative therapies, adoptive cell therapy (ACT) has emerged as a promising cancer treatment in recent years. The limited clinical applications of ACT, despite its advantages over standard-of-care therapies, can be attributed to (i) time-consuming and cost-intensive procedures to screen for potent anti-tumor immune cells and the corresponding targets, (ii) difficulties to translate in-vitro and animal-derived in-vivo efficacies to clinical efficacy in humans, and (iii) the lack of systemic methods for the safety assessment of ACT. Suitable experimental models and testing platforms have the potential to accelerate the development of ACT. Immunocompetent microphysiological systems (iMPS) are microfluidic platforms that enable complex interactions of advanced tissue models with different immune cell types, bridging the gap between in-vitro and in-vivo studies. Here, we present a proof-of-concept iMPS that supports a triple culture of three-dimensional (3D) colorectal tumor microtissues, 3D cardiac microtissues, and human-derived natural killer (NK) cells in the same microfluidic network. Different aspects of tumor-NK cell interactions were characterized using this iMPS including: (i) direct interaction and NK cell-mediated tumor killing, (ii) the development of an inflammatory milieu through enrichment of soluble pro-inflammatory chemokines and cytokines, and (iii) secondary effects on healthy cardiac microtissues. We found a specific NK cell-mediated tumor-killing activity and elevated levels of tumor- and NK cell-derived chemokines and cytokines, indicating crosstalk and development of an inflammatory milieu. While viability and morphological integrity of cardiac microtissues remained mostly unaffected, we were able to detect alterations in their beating behavior, which shows the potential of iMPS for both, efficacy and early safety testing of new candidate ACTs.


Assuntos
Bioensaio/métodos , Técnicas de Cultura de Células em Três Dimensões/métodos , Imunoterapia Adotiva , Células Matadoras Naturais/transplante , Neoplasias/terapia , Bioensaio/instrumentação , Técnicas de Cultura de Células em Três Dimensões/instrumentação , Linhagem Celular , Separação Celular , Feminino , Sangue Fetal , Voluntários Saudáveis , Humanos , Células-Tronco Pluripotentes Induzidas , Microscopia Intravital , Células Matadoras Naturais/imunologia , Dispositivos Lab-On-A-Chip , Masculino , Miócitos Cardíacos , Neoplasias/imunologia , Neoplasias/patologia , Cultura Primária de Células , Estudo de Prova de Conceito
6.
Front Pharmacol ; 12: 785851, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35342386

RESUMO

Understanding the pharmacokinetic/pharmacodynamic (PK/PD)-relationship of a drug candidate is key to determine effective, yet safe treatment regimens for patients. However, current testing strategies are inefficient in characterizing in vivo responses to fluctuating drug concentrations during multi-day treatment cycles. Methods based on animal models are resource-intensive and require time, while traditional in vitro cell-culturing methods usually do not provide temporally-resolved information on the effects of in vivo-like drug exposure scenarios. To address this issue, we developed a microfluidic system to 1) culture arrays of three-dimensional spheroids in vitro, to 2) apply specific dynamic drug exposure profiles, and to 3) in-situ analyze spheroid growth and the invoked drug effects in 3D by means of 2-photon microscopy at tissue and single-cell level. Spheroids of fluorescently-labeled T-47D breast cancer cells were monitored under perfusion-culture conditions at short time intervals over three days and exposed to either three 24 h-PK-cycles or a dose-matched constant concentration of the phosphatidylinositol 3-kinase inhibitor BYL719. While the overall efficacy of the two treatment regimens was similar, spheroids exposed to the PK profile displayed cycle-dependent oscillations between regression and regrowth. Spheroids treated with a constant BYL719 concentration regressed at a steady, albeit slower rate. At a single-cell level, the cell density in BYL719-treated spheroids oscillated in a concentration-dependent manner. Our system represents a versatile tool for in-depth preclinical characterization of PK/PD parameters, as it enables an evaluation of drug efficacy and/or toxicity under realistic exposure conditions.

7.
Adv Biosyst ; 4(11): e2000079, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33073544

RESUMO

Drug-drug interactions (DDIs) occur when the pharmacological activity of one drug is altered by a second drug. As multimorbidity and polypharmacotherapy are becoming more common due to the increasing age of the population, the risk of DDIs is massively increasing. Therefore, in vitro testing methods are needed to capture such multiorgan events. Here, a scalable, gravity-driven microfluidic system featuring 3D microtissues (MTs) that represent different organs for the prediction of drug-drug interactions is used. Human liver microtissues (hLiMTs) are combined with tumor microtissues (TuMTs) and treated with drug combinations that are known to cause DDIs in vivo. The testing system is able to capture and quantify DDIs upon co-administration of the anticancer prodrugs cyclophosphamide or ifosfamide with the antiretroviral drug ritonavir. Dosage of ritonavir inhibits hepatic metabolization of the two prodrugs to different extents and decreases their efficacy in acting on TuMTs. The flexible MT compartment design of the system, the use of polystyrene as chip material, and the assembly of several chips in stackable plates offer the potential to significantly advance preclinical substance testing. The possibility of testing a broad variety of drug combinations to identify possible DDIs will improve the drug development process and increase patient safety.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Interações Medicamentosas , Técnicas Analíticas Microfluídicas , Análise Serial de Tecidos/métodos , Técnicas de Cultura de Tecidos/métodos , Sobrevivência Celular/efeitos dos fármacos , Células HCT116 , Inibidores da Protease de HIV/farmacologia , Humanos , Fígado/citologia , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos , Ritonavir/farmacologia
8.
Artigo em Inglês | MEDLINE | ID: mdl-31001529

RESUMO

In vitro screening methods for compound efficacy and toxicity to date mostly include cell or tissue exposure to preset constant compound concentrations over a defined testing period. Such concentration profiles, however, do not represent realistic in vivo situations after substance uptake. Absorption, distribution, metabolism and excretion of administered substances in an organism or human body entail gradually changing pharmacokinetic concentration profiles. As concentration profile dynamics can influence drug effects on the target tissues, it is important to be able to reproduce realistic concentration profiles in in vitro systems. We present a novel design that can be integrated in tubing-free, microfluidic culture chips. These chips are actuated by tilting so that gravity-driven flow and perfusion of culture chambers can be established between reservoirs at both ends of a microfluidic channel. The design enables the realization of in vivo-like substance exposure scenarios. Compound gradients are generated through an asymmetric Y-junction of channels with different hydrodynamic resistances. Six microtissues (MTs) can be cultured and exposed in compartments along the channel. Changes of the chip design or operation parameters enable to alter the dosing profile over a large range. Modulation of, e.g., the tilting angle, changes the slope of the dosing curves, so that concentration curves can be attained that resemble the pharmacokinetic characteristics of common substances in a human body. Human colorectal cancer (HCT 116) MTs were exposed to both, gradually decreasing and constant concentrations of Staurosporine. Measurements of apoptosis induction and viability after 5 h and 24 h showed different short- and long-term responses of the MTs to dynamic and linear dosing regimes.

9.
SLAS Technol ; 24(1): 79-95, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30289726

RESUMO

Microphysiological systems hold the promise to increase the predictive and translational power of in vitro substance testing owing to their faithful recapitulation of human physiology. However, the implementation of academic developments in industrial settings remains challenging. We present an injection-molded microfluidic microtissue (MT) culture chip that features two channels with 10 MT compartments each and that was designed in compliance with microtiter plate standard formats. Polystyrene as a chip material enables reliable, large-scale production and precise control over experimental conditions due to low adsorption or absorption of small, hydrophobic molecules at or into the plastic material in comparison with predecessor chips made of polydimethylsiloxane. The chip is operated by tilting, which actuates gravity-driven flow between reservoirs at both ends of every channel, so that the system does not require external tubing or pumps. The flow rate can be modulated by adjusting the tilting angle on demand. The top-open design of the MT compartment enables efficient MT loading using standard or advanced pipetting equipment, ensures oxygen availability in the chip, and allows for high-resolution imaging. Every channel can be loaded with up to 10 identical or different MTs, as demonstrated by culturing liver and tumor MTs in the same medium channel on the chip.


Assuntos
Microfluídica/instrumentação , Microfluídica/métodos , Técnicas de Cultura de Órgãos/métodos , Técnicas de Cultura de Tecidos/métodos , Humanos , Poliestirenos , Alicerces Teciduais
10.
Adv Biosyst ; 3(5): e1900018, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-32627410

RESUMO

Studying and understanding of tissue and disease mechanisms largely depend on the availability of suitable and representative biological model systems. These model systems should be carefully engineered and faithfully reproduce the biological system of interest to understand physiological effects, pharmacokinetics, and toxicity to better identify new drug compounds. By relying on microfluidics, microphysiological systems (MPSs) enable the precise control of culturing conditions and connections of advanced in vitro 3D organ models that better reproduce in vivo environments. This review focuses on transferable in vitro organ models and integrated MPSs that host these transferable biological units and enable interactions between different tissue types. Interchangeable and transferrable in vitro organ models allow for independent quality control of the biological model before system assembly and building MPS assays on demand. Due to the complexity and different maturation times of individual in vitro tissues, off-chip production and quality control entail improved stability and reproducibility of the systems and results, which is important for large-scale adoption of the technology. Lastly, the technical and biological challenges and open issues for realizing and implementing integrated MPSs with transferable in vitro organ models are discussed.


Assuntos
Dispositivos Lab-On-A-Chip , Procedimentos Analíticos em Microchip , Modelos Biológicos , Engenharia Tecidual , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA