Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Biomed Eng ; 8(8): 941-962, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39187664

RESUMO

Microphysiological systems (MPSs) are cellular models that replicate aspects of organ and tissue functions in vitro. In contrast with conventional cell cultures, MPSs often provide physiological mechanical cues to cells, include fluid flow and can be interlinked (hence, they are often referred to as microfluidic tissue chips or organs-on-chips). Here, by means of examples of MPSs of the vascular system, intestine, brain and heart, we advocate for the development of standards that allow for comparisons of quantitative physiological features in MPSs and humans. Such standards should ensure that the in vivo relevance and predictive value of MPSs can be properly assessed as fit-for-purpose in specific applications, such as the assessment of drug toxicity, the identification of therapeutics or the understanding of human physiology or disease. Specifically, we distinguish designed features, which can be controlled via the design of the MPS, from emergent features, which describe cellular function, and propose methods for improving MPSs with readouts and sensors for the quantitative monitoring of complex physiology towards enabling wider end-user adoption and regulatory acceptance.


Assuntos
Dispositivos Lab-On-A-Chip , Humanos , Animais , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos , Microfluídica/métodos , Modelos Biológicos , Encéfalo/fisiologia , Desenho de Equipamento , Sistemas Microfisiológicos
2.
Lab Chip ; 23(1): 168-181, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36484766

RESUMO

Three-dimensional (3D) blood vessels-on-a-chip (VoC) models integrate the biological complexity of vessel walls with dynamic microenvironmental cues, such as wall shear stress (WSS) and circumferential strain (CS). However, these parameters are difficult to control and are often poorly reproducible due to the high intrinsic diameter variation of individual 3D-VoCs. As a result, the throughput of current 3D systems is one-channel-at-a-time. Here, we developed a fluidic circuit board (FCB) for simultaneous perfusion of up to twelve 3D-VoCs using a single set of control parameters. By designing the internal hydraulic resistances in the FCB appropriately, it was possible to provide a pre-set WSS to all connected 3D-VoCs, despite significant variation in lumen diameters. Using this FCB, we found that variation of CS or WSS induce morphological changes to human induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) and conclude that control of these parameters using a FCB is necessary to study 3D-VOCs.


Assuntos
Células Endoteliais , Células-Tronco Pluripotentes Induzidas , Humanos , Perfusão , Dispositivos Lab-On-A-Chip , Estresse Mecânico
3.
Mater Today Bio ; 17: 100475, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36388452

RESUMO

Recent advances in microfluidic engineering allow the creation of microenvironments in which human cells can be cultured under (patho-)physiological conditions with greater reality than standard plastic tissue culture plates. Microfluidic devices, also called Organs-on-Chip (OoC), allow complex engineering of the cellular compartment, yielding designs in which microfluidic flow can be precisely controlled. However, it is important that cellular physiology is not only controlled but can also be monitored in these devices. Here, we integrated oxygen and pH sensors into microfluidics, allowing close monitoring of the extracellular flux from the cells, enabling constant assessment of features such as glycolysis and mitochondrial oxidative phosphorylation in situ. Using human-induced pluripotent stem cells (hiPSCs) as an exemplar of a highly metabolic and relatively challenging cell type to maintain, we showed that monitoring the extracellular environment allowed rapid optimization of the seeding protocol. Based on the measurements, we implemented earlier and more frequent media refreshment to counteract the rapid acidification and depletion of oxygen. The integrated sensors showed that hiPSCs in the devices exhibited mitochondrial and glycolytic capacity similar to that measured with the Seahorse extracellular flux system, the most widely used standard for these types of assays in conventional cell culture. Under both conditions, hiPSCs showed greater reliance on glycolysis than mitochondrial OXPHOS and the absolute values obtained were similar. These results thus pave the way for the assessment of cell metabolism in situ under conditions of fluidic flow with the same precision and relevance as current standard static cell cultures.

4.
Micromachines (Basel) ; 13(8)2022 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-36014281

RESUMO

Organ-on-chip (OoC) devices are increasingly used to mimic the tissue microenvironment of cells in intact organs. This includes microchannels to mimic, for example, fluidic flow through blood vessels. Present methods for controlling microfluidic flow in these systems rely on gravity, rocker systems or external pressure pumps. For many purposes, pressure pumps give the most consistent flow profiles, but they are not well-suited for high throughput as might be required for testing drug responses. Here, we describe a method which allows for multiplexing of microfluidic channels in OoC devices plus the accompanying custom software necessary to run the system. Moreover, we show the approach is also suitable for recirculation of culture medium, an essential cost consideration when expensive culture reagents are used and are not "spent" through uptake by the cells during transient unidirectional flow.

5.
Micromachines (Basel) ; 13(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35056214

RESUMO

Organ-on-a-chip (OoC) and microfluidic devices are conventionally produced using microfabrication procedures that require cleanrooms, silicon wafers, and photomasks. The prototyping stage often requires multiple iterations of design steps. A simplified prototyping process could therefore offer major advantages. Here, we describe a rapid and cleanroom-free microfabrication method using maskless photolithography. The approach utilizes a commercial digital micromirror device (DMD)-based setup using 375 nm UV light for backside exposure of an epoxy-based negative photoresist (SU-8) on glass coverslips. We show that microstructures of various geometries and dimensions, microgrooves, and microchannels of different heights can be fabricated. New SU-8 molds and soft lithography-based polydimethylsiloxane (PDMS) chips can thus be produced within hours. We further show that backside UV exposure and grayscale photolithography allow structures of different heights or structures with height gradients to be developed using a single-step fabrication process. Using this approach: (1) digital photomasks can be designed, projected, and quickly adjusted if needed; and (2) SU-8 molds can be fabricated without cleanroom availability, which in turn (3) reduces microfabrication time and costs and (4) expedites prototyping of new OoC devices.

6.
APL Bioeng ; 3(2): 026105, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31263797

RESUMO

Blood vessel models are increasingly recognized to have value in understanding disease and drug discovery. However, continued improvements are required to more accurately reflect human vessel physiology. Realistic three-dimensional (3D) in vitro cultures of human vascular cells inside microfluidic chips, or vessels-on-chips (VoC), could contribute to this since they can recapitulate aspects of the in vivo microenvironment by including mechanical stimuli such as shear stress. Here, we used human induced pluripotent stem cells as a source of endothelial cells (hiPSC-ECs), in combination with a technique called viscous finger patterning (VFP) toward this goal. We optimized VFP to create hollow structures in collagen I extracellular-matrix inside microfluidic chips. The lumen formation success rate was over 90% and the resulting cellularized lumens had a consistent diameter over their full length, averaging 336 ± 15 µm. Importantly, hiPSC-ECs cultured in these 3D microphysiological systems formed stable and viable vascular structures within 48 h. Furthermore, this system could support coculture of hiPSC-ECs with primary human brain vascular pericytes, demonstrating their ability to accommodate biologically relevant combinations of multiple vascular cell types. Our protocol for VFP is more robust than previously published methods with respect to success rates and reproducibility of the diameter between- and within channels. This, in combination with the ease of preparation, makes hiPSC-EC based VoC a low-cost platform for future studies in personalized disease modeling.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA