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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nat Biomed Eng ; 6(11): 1236-1247, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35739419

RESUMEN

Environmental enteric dysfunction (EED)-a chronic inflammatory condition of the intestine-is characterized by villus blunting, compromised intestinal barrier function and reduced nutrient absorption. Here we show that essential genotypic and phenotypic features of EED-associated intestinal injury can be reconstituted in a human intestine-on-a-chip lined by organoid-derived intestinal epithelial cells from patients with EED and cultured in nutrient-deficient medium lacking niacinamide and tryptophan. Exposure of the organ chip to such nutritional deficiencies resulted in congruent changes in six of the top ten upregulated genes that were comparable to changes seen in samples from patients with EED. Chips lined with healthy epithelium or with EED epithelium exposed to nutritional deficiencies resulted in severe villus blunting and barrier dysfunction, and in the impairment of fatty acid uptake and amino acid transport; and the chips with EED epithelium exhibited heightened secretion of inflammatory cytokines. The organ-chip model of EED-associated intestinal injury may facilitate the analysis of the molecular, genetic and nutritional bases of the disease and the testing of candidate therapeutics for it.


Asunto(s)
Enfermedades Intestinales , Desnutrición , Humanos , Dispositivos Laboratorio en un Chip , Intestinos , Intestino Delgado/metabolismo , Desnutrición/metabolismo
2.
Front Pharmacol ; 12: 718484, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34759819

RESUMEN

Many patients infected with coronaviruses, such as SARS-CoV-2 and NL63 that use ACE2 receptors to infect cells, exhibit gastrointestinal symptoms and viral proteins are found in the human gastrointestinal tract, yet little is known about the inflammatory and pathological effects of coronavirus infection on the human intestine. Here, we used a human intestine-on-a-chip (Intestine Chip) microfluidic culture device lined by patient organoid-derived intestinal epithelium interfaced with human vascular endothelium to study host cellular and inflammatory responses to infection with NL63 coronavirus. These organoid-derived intestinal epithelial cells dramatically increased their ACE2 protein levels when cultured under flow in the presence of peristalsis-like mechanical deformations in the Intestine Chips compared to when cultured statically as organoids or in Transwell inserts. Infection of the intestinal epithelium with NL63 on-chip led to inflammation of the endothelium as demonstrated by loss of barrier function, increased cytokine production, and recruitment of circulating peripheral blood mononuclear cells (PBMCs). Treatment of NL63 infected chips with the approved protease inhibitor drug, nafamostat, inhibited viral entry and resulted in a reduction in both viral load and cytokine secretion, whereas remdesivir, one of the few drugs approved for COVID19 patients, was not found to be effective and it also was toxic to the endothelium. This model of intestinal infection was also used to test the effects of other drugs that have been proposed for potential repurposing against SARS-CoV-2. Taken together, these data suggest that the human Intestine Chip might be useful as a human preclinical model for studying coronavirus related pathology as well as for testing of potential anti-viral or anti-inflammatory therapeutics.

3.
Nat Biomed Eng ; 4(4): 421-436, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31988459

RESUMEN

Analyses of drug pharmacokinetics (PKs) and pharmacodynamics (PDs) performed in animals are often not predictive of drug PKs and PDs in humans, and in vitro PK and PD modelling does not provide quantitative PK parameters. Here, we show that physiological PK modelling of first-pass drug absorption, metabolism and excretion in humans-using computationally scaled data from multiple fluidically linked two-channel organ chips-predicts PK parameters for orally administered nicotine (using gut, liver and kidney chips) and for intravenously injected cisplatin (using coupled bone marrow, liver and kidney chips). The chips are linked through sequential robotic liquid transfers of a common blood substitute by their endothelium-lined channels (as reported by Novak et al. in an associated Article) and share an arteriovenous fluid-mixing reservoir. We also show that predictions of cisplatin PDs match previously reported patient data. The quantitative in-vitro-to-in-vivo translation of PK and PD parameters and the prediction of drug absorption, distribution, metabolism, excretion and toxicity through fluidically coupled organ chips may improve the design of drug-administration regimens for phase-I clinical trials.


Asunto(s)
Dispositivos Laboratorio en un Chip , Microfluídica/métodos , Preparaciones Farmacéuticas , Farmacocinética , Animales , Cisplatino/farmacocinética , Diseño de Fármacos , Humanos , Técnicas In Vitro , Hígado/metabolismo , Microfluídica/instrumentación , Modelos Biológicos , Nicotina/farmacocinética , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/metabolismo
4.
Nat Biomed Eng ; 4(4): 407-420, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31988458

RESUMEN

Organ chips can recapitulate organ-level (patho)physiology, yet pharmacokinetic and pharmacodynamic analyses require multi-organ systems linked by vascular perfusion. Here, we describe an 'interrogator' that employs liquid-handling robotics, custom software and an integrated mobile microscope for the automated culture, perfusion, medium addition, fluidic linking, sample collection and in situ microscopy imaging of up to ten organ chips inside a standard tissue-culture incubator. The robotic interrogator maintained the viability and organ-specific functions of eight vascularized, two-channel organ chips (intestine, liver, kidney, heart, lung, skin, blood-brain barrier and brain) for 3 weeks in culture when intermittently fluidically coupled via a common blood substitute through their reservoirs of medium and endothelium-lined vascular channels. We used the robotic interrogator and a physiological multicompartmental reduced-order model of the experimental system to quantitatively predict the distribution of an inulin tracer perfused through the multi-organ human-body-on-chips. The automated culture system enables the imaging of cells in the organ chips and the repeated sampling of both the vascular and interstitial compartments without compromising fluidic coupling.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Dispositivos Laboratorio en un Chip , Microfluídica/métodos , Robótica/métodos , Barrera Hematoencefálica , Encéfalo , Calibración , Técnicas de Cultivo de Célula/instrumentación , Diseño de Equipo , Corazón , Humanos , Intestinos , Riñón , Hígado , Pulmón , Robótica/instrumentación , Piel
5.
Nat Biomed Eng ; 3(7): 520-531, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31086325

RESUMEN

The diverse bacterial populations that comprise the commensal microbiome of the human intestine play a central role in health and disease. A method that sustains complex microbial communities in direct contact with living human intestinal cells and their overlying mucus layer in vitro would thus enable the investigation of host-microbiome interactions. Here, we show the extended coculture of living human intestinal epithelium with stable communities of aerobic and anaerobic human gut microbiota, using a microfluidic intestine-on-a-chip that permits the control and real-time assessment of physiologically relevant oxygen gradients. When compared to aerobic coculture conditions, the establishment of a transluminal hypoxia gradient in the chip increased intestinal barrier function and sustained a physiologically relevant level of microbial diversity, consisting of over 200 unique operational taxonomic units from 11 different genera and an abundance of obligate anaerobic bacteria, with ratios of Firmicutes and Bacteroidetes similar to those observed in human faeces. The intestine-on-a-chip may serve as a discovery tool for the development of microbiome-related therapeutics, probiotics and nutraceuticals.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/microbiología , Dispositivos Laboratorio en un Chip , Microbiota/fisiología , Técnicas Analíticas Microfluídicas/métodos , Anaerobiosis , Bacterias/clasificación , Bacterias/crecimiento & desarrollo , Bacteroidetes , Biodiversidad , Células CACO-2 , Células Epiteliales , Heces/microbiología , Firmicutes , Interacciones Microbiota-Huesped/fisiología , Humanos , Hipoxia , Técnicas In Vitro , Moco , Oxígeno
6.
Nat Biomed Eng ; 3(7): 583, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31213704

RESUMEN

In the version of this Article originally published, the authors mistakenly cited Fig. 5d in the sentence beginning 'Importantly, the microbiome cultured in these primary Intestine Chips...'; the correct citation is Supplementary Table 2. This has now been amended.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA