Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Adv Drug Deliv Rev ; 191: 114542, 2022 12.
Article in English | MEDLINE | ID: mdl-36179916

ABSTRACT

The surfaces of human internal organs are lined by a mucus layer that ensures symbiotic relationships with commensal microbiome while protecting against potentially injurious environmental chemicals, toxins, and pathogens, and disruption of this layer can contribute to disease development. Studying mucus biology has been challenging due to the lack of physiologically relevant human in vitro models. Here we review recent progress that has been made in the development of human organ-on-a-chip microfluidic culture models that reconstitute epithelial tissue barriers and physiologically relevant mucus layers with a focus on lung, colon, small intestine, cervix and vagina. These organ-on-a-chip models that incorporate dynamic fluid flow, air-liquid interfaces, and physiologically relevant mechanical cues can be used to study mucus composition, mechanics, and structure, as well as investigate its contributions to human health and disease with a level of biomimicry not possible in the past.


Subject(s)
Models, Biological , Mucus , Humans , Colon , Lab-On-A-Chip Devices , Microbiota , Microfluidics , Mucus/physiology
2.
Nat Biomed Eng ; 6(11): 1236-1247, 2022 11.
Article in English | MEDLINE | ID: mdl-35739419

ABSTRACT

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.


Subject(s)
Intestinal Diseases , Malnutrition , Humans , Lab-On-A-Chip Devices , Intestines , Intestine, Small/metabolism , Malnutrition/metabolism
3.
Methods Mol Biol ; 2373: 69-85, 2022.
Article in English | MEDLINE | ID: mdl-34520007

ABSTRACT

It is impossible to analyze human-specific host-microbiome interactions using animal models and existing in vitro methods fail to support survival of human cells in direct contact with complex living microbiota for extended times. Here we describe a protocol for culturing human organ-on-a-chip (Organ Chip) microfluidic devices lined by human patient-derived primary intestinal epithelium in the presence of a physiologically relevant transluminal hypoxia gradient that enables their coculture with hundreds of different living aerobic and anaerobic bacteria found within the human gut microbiome. This protocol can be adapted to provide different levels of oxygen tension to facilitate coculturing of microbiome from different regions of gastrointestinal tract, and the same system can be applied with any other type of Organ Chip. This method can help to provide further insight into the host-microbiome interactions that contribute to human health and disease, enable discovery of new microbiome-related diagnostics and therapeutics, and provide a novel approach to advanced personalized medicine.


Subject(s)
Gastrointestinal Microbiome , Anaerobiosis , Animals , Humans , Intestinal Mucosa/metabolism , Lab-On-A-Chip Devices
4.
Front Pharmacol ; 12: 718484, 2021.
Article in English | MEDLINE | ID: mdl-34759819

ABSTRACT

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.

5.
Nat Biomed Eng ; 3(7): 583, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31213704

ABSTRACT

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.

6.
Nat Biomed Eng ; 3(7): 520-531, 2019 07.
Article in English | MEDLINE | ID: mdl-31086325

ABSTRACT

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.


Subject(s)
Cell Culture Techniques/methods , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/microbiology , Lab-On-A-Chip Devices , Microbiota/physiology , Microfluidic Analytical Techniques/methods , Anaerobiosis , Bacteria/classification , Bacteria/growth & development , Bacteroidetes , Biodiversity , Caco-2 Cells , Epithelial Cells , Feces/microbiology , Firmicutes , Host Microbial Interactions/physiology , Humans , Hypoxia , In Vitro Techniques , Mucus , Oxygen
7.
Lab Chip ; 19(3): 452-463, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30632575

ABSTRACT

Here, we describe methods for combining impedance spectroscopy measurements with electrical simulation to reveal transepithelial barrier function and tissue structure of human intestinal epithelium cultured inside an organ-on-chip microfluidic culture device. When performing impedance spectroscopy measurements, electrical simulation enabled normalization of cell layer resistance of epithelium cultured statically in a gut-on-a-chip, which enabled determination of transepithelial electrical resistance (TEER) values that can be compared across device platforms. During culture under dynamic flow, the formation of intestinal villi was accompanied by characteristic changes in impedance spectra both measured experimentally and verified with simulation, and we demonstrate that changes in cell layer capacitance may serve as measures of villi differentiation. This method for combining impedance spectroscopy with simulation can be adapted to better monitor cell layer characteristics within any organ-on-chip in vitro and to enable direct quantitative TEER comparisons between organ-on-chip platforms which should help to advance research on organ function.


Subject(s)
Dielectric Spectroscopy/instrumentation , Intestinal Mucosa/cytology , Lab-On-A-Chip Devices , Caco-2 Cells , Electric Stimulation , Equipment Design , Humans , Intestinal Mucosa/metabolism
8.
Cell Mol Gastroenterol Hepatol ; 5(4): 659-668, 2018.
Article in English | MEDLINE | ID: mdl-29713674

ABSTRACT

Microfluidic organ-on-a-chip models of human intestine have been developed and used to study intestinal physiology and pathophysiology. In this article, we review this field and describe how microfluidic Intestine Chips offer new capabilities not possible with conventional culture systems or organoid cultures, including the ability to analyze contributions of individual cellular, chemical, and physical control parameters one-at-a-time; to coculture human intestinal cells with commensal microbiome for extended times; and to create human-relevant disease models. We also discuss potential future applications of human Intestine Chips, including how they might be used for drug development and personalized medicine.

9.
Sci Rep ; 8(1): 2871, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29440725

ABSTRACT

Here we describe a method for fabricating a primary human Small Intestine-on-a-Chip (Intestine Chip) containing epithelial cells isolated from healthy regions of intestinal biopsies. The primary epithelial cells are expanded as 3D organoids, dissociated, and cultured on a porous membrane within a microfluidic device with human intestinal microvascular endothelium cultured in a parallel microchannel under flow and cyclic deformation. In the Intestine Chip, the epithelium forms villi-like projections lined by polarized epithelial cells that undergo multi-lineage differentiation similar to that of intestinal organoids, however, these cells expose their apical surfaces to an open lumen and interface with endothelium. Transcriptomic analysis also indicates that the Intestine Chip more closely mimics whole human duodenum in vivo when compared to the duodenal organoids used to create the chips. Because fluids flowing through the lumen of the Intestine Chip can be collected continuously, sequential analysis of fluid samples can be used to quantify nutrient digestion, mucus secretion and establishment of intestinal barrier function over a period of multiple days in vitro. The Intestine Chip therefore may be useful as a research tool for applications where normal intestinal function is crucial, including studies of metabolism, nutrition, infection, and drug pharmacokinetics, as well as personalized medicine.


Subject(s)
Intestine, Small/cytology , Lab-On-A-Chip Devices , Organoids/cytology , Biopsy , Cell Proliferation , Epithelial Cells/cytology , Humans
10.
PLoS One ; 13(2): e0192265, 2018.
Article in English | MEDLINE | ID: mdl-29447224

ABSTRACT

Successfully cryopreserving cells adhered to a substrate would facilitate the growth of a vital confluent cell culture after thawing while dramatically shortening the post-thaw culturing time. Herein we propose a controlled slow cooling method combining initial directional freezing followed by gradual cooling down to -80°C for robust preservation of cell monolayers adherent to a substrate. Using computer controlled cryostages we examined the effect of cooling rates and dimethylsulfoxide (DMSO) concentration on cell survival and established an optimal cryopreservation protocol. Experimental results show the highest post-thawing viability for directional ice growth at a speed of 30 µm/sec (equivalent to freezing rate of 3.8°C/min), followed by gradual cooling of the sample with decreasing rate of 0.5°C/min. Efficient cryopreservation of three widely used epithelial cell lines: IEC-18, HeLa, and Caco-2, provides proof-of-concept support for this new freezing protocol applied to adherent cells. This method is highly reproducible, significantly increases the post-thaw cell viability and can be readily applied for cryopreservation of cellular cultures in microfluidic devices.


Subject(s)
Cell Adhesion , Cryopreservation , Freezing , Caco-2 Cells , Cell Survival , Humans
11.
Pediatr Neonatol ; 59(5): 464-473, 2018 10.
Article in English | MEDLINE | ID: mdl-29276042

ABSTRACT

BACKGROUND & AIMS: Necrotizing Enterocolitis (NEC) is a severe inflammatory disorder of the intestine endangering the health and survival of preterm infants. It is well established that the gut barrier is severely damaged in NEC patients, nonetheless an in depth investigation of modifications at the transcriptional and translational levels of tight junction genes and proteins during NEC are still missing. The aim of this study was to investigate changes in the expression of tight junctions and other associated proteins during NEC and determine their correlation to the disease severity. METHODS: We examined intestinal specimens from six NEC patients and compared them with six control specimens from patients that underwent surgeries for reasons other than NEC. The expression of genes was analyzed by real time PCR and protein expression by immunohistochemistry. RESULTS: The tight junction genes ZO-1, occludin, cingulin and claudin-4 were significantly down regulated in NEC. Furthermore TLR4, BAX and SIRT1 genes were found to be significantly down regulated while HIF-1A showed a trend of up regulation in NEC patients. These changes were found to correlate with the severity of the disease. Additionally we demonstrated in an ex-vivo model that hypoxic conditions initiated a destructive process of the epithelial barrier. We also showed that the expression of the tight junction proteins ZO-1 and occludin were significantly down regulated in NEC specimens. CONCLUSIONS: The expression of tight junction proteins and their encoding genes are significantly altered in NEC. We surmise that SIRT1 and HIF-1A may play a role in controlling these effects.


Subject(s)
Enterocolitis, Necrotizing/metabolism , Intestinal Mucosa/metabolism , Tight Junctions/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Infant , Infant, Newborn , Infant, Premature , Male , Occludin/genetics , Sirtuin 1/physiology , Zonula Occludens-1 Protein/genetics
12.
J Cell Physiol ; 232(2): 381-390, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27191060

ABSTRACT

Necrotizing Enterocolitis (NEC) is a severe inflammatory disorder leading to high morbidity and mortality rates. A growing body of evidence demonstrate the key role of the Toll like receptor 4 (TLR4) in NEC. This membranal receptor recognizes lipopolysaccharides (LPS) from the bacterial wall and triggers an inflammatory response. The aim of the present study was to elucidate the effect of LPS on paracellular permeability known to be severely affected in NEC. IEC-18 cells were treated with LPS and the effects on morphology, paracellular permeability and their associated gene and protein expressions were measured. Our results show that LPS down regulated the expression of occludin and ZO-1 mRNAs while up regulating Cdkn1a. In addition LPS caused a significant increase in paracellular permeability and epithelial barrier damage. Finally ZO-1 protein was found to be spatially disarrayed in the intercellular junctions in response to LPS. We conclude that LPS adversely affected the functionality of the intestinal epithelial barrier suggesting a new mechanism by which bacterial infection may contribute to the development of NEC. J. Cell. Physiol. 232: 381-390, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Membrane Permeability/drug effects , Epithelial Cells/cytology , Intestines/cytology , Lipopolysaccharides/pharmacology , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/ultrastructure , Gene Expression Regulation/drug effects , Humans , NF-kappa B/metabolism , Signal Transduction/drug effects , Surface Plasmon Resonance , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Toll-Like Receptor 4/metabolism , Zonula Occludens-1 Protein/metabolism
13.
Pediatr Res ; 77(5): 649-55, 2015 May.
Article in English | MEDLINE | ID: mdl-25665057

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC), a common intestinal disease affecting premature infants, is a major cause of morbidity and mortality. Previous reports indicate an upregulation of intestinal matrix metalloproteinases (MMPs) activity that may play key roles on the higher permeability of the intestinal barrier, typical to NEC. Recently, TIMP-1, a natural inhibitor of MMP's, was found to be over expressed in preterm human breast milk (HBM). Previous studies have shown that infants fed with HBM have a significant reduction in the incidence of NEC. The aim of the present study was to investigate the possible role that TIMP-1 may play on the maintenance of tight junctions and therefore the gut barrier integrity. METHODS: Timp-1-treated Caco-2 intestinal cells were tested for MMP-2 enzymatic activity and cell junction integrity. RESULTS: TIMP-1 inhibited MMP-2 activity, which induced a significant increase in the expression of occludin but not of claudin-4. TIMP-1 did not affect apoptosis. CONCLUSION: One of the putative mechanisms associated with HBM protection against NEC is mediated by TIMP-1, which downregulates MMP-2 activity, inhibits the degradation of occluding, and preserves tight junctions and gut barrier integrity.


Subject(s)
Enterocolitis, Necrotizing/metabolism , Gene Expression Regulation , Intestines/pathology , Matrix Metalloproteinase 2/metabolism , Occludin/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Caco-2 Cells , Humans , Intestinal Mucosa/metabolism , Milk, Human/metabolism , Permeability , Recombinant Proteins/metabolism , Tight Junctions/pathology
14.
J Biomed Opt ; 19(11): 111608, 2014.
Article in English | MEDLINE | ID: mdl-24972266

ABSTRACT

We report a label-free infrared surface plasmon biosensor with a double-chamber flow cell for continuous monitoring of morphological changes in cell culture exposed to various stimuli. In this technique, the monolayer of cultured cells is divided into two halves by a barrier, allowing the treatment of one half while the other serves as control. We demonstrate the advantages of this setup in test experiments that track kinetics of the IEC-18 cell layer response to variations in extracellular Ca2+ concentration. The sensitivity of the presented method was found to be an order of magnitude higher compared to the single-chamber biosensor.


Subject(s)
Biosensing Techniques/instrumentation , Cell Culture Techniques/instrumentation , Surface Plasmon Resonance/instrumentation , Animals , Biosensing Techniques/methods , Calcium/pharmacology , Cell Culture Techniques/methods , Cell Line , Cell Shape/drug effects , Rats , Refractometry , Surface Plasmon Resonance/methods
15.
J Biomed Opt ; 17(8): 081409-1, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23224170

ABSTRACT

Cell morphology is often used as a valuable indicator of the physical condition and general status of living cells. We demonstrate a noninvasive method for morphological characterization of adherent cells. We measure infrared reflectivity spectrum at oblique angle from living cells cultured on thin Au film, and utilize the unique properties of the confined infrared waves (i.e., surface plasmon and guided modes) traveling inside the cell layer. The propagation of these waves strongly depends on cell morphology and connectivity. By tracking the resonant wavelength and attenuation of the surface plasmon and guided modes we measure the kinetics of various cellular processes such as (i) cell attachment and spreading on different substrata, (ii) modulation of the outer cell membrane with chlorpromazine, and (iii) formation of intercellular junctions associated with progressive cell polarization. Our method enables monitoring of submicron variations in cell layer morphology in real-time, and in the label-free manner.


Subject(s)
Biosensing Techniques/instrumentation , Cell Adhesion/physiology , Cell Polarity/physiology , Cell Separation/instrumentation , Flow Cytometry/instrumentation , Spectrophotometry, Infrared/instrumentation , Surface Plasmon Resonance/instrumentation , Animals , Cell Size , Dogs , Equipment Design , Equipment Failure Analysis , Madin Darby Canine Kidney Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...