Please use this identifier to cite or link to this item: https://doi.org/10.1371/journal.pone.0197101
DC FieldValue
dc.titleA multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies
dc.contributor.authorTan H.-Y.
dc.contributor.authorTrier S.
dc.contributor.authorRahbek U.L.
dc.contributor.authorDufva M.
dc.contributor.authorKutter J.P.
dc.contributor.authorAndresen T.L.
dc.date.accessioned2019-11-01T08:15:25Z
dc.date.available2019-11-01T08:15:25Z
dc.date.issued2018
dc.identifier.citationTan H.-Y., Trier S., Rahbek U.L., Dufva M., Kutter J.P., Andresen T.L. (2018). A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies. PLoS ONE 13 (5) : e0197101. ScholarBank@NUS Repository. https://doi.org/10.1371/journal.pone.0197101
dc.identifier.issn19326203
dc.identifier.urihttps://scholarbank.nus.edu.sg/handle/10635/161229
dc.description.abstractThis paper presents the design and fabrication of a multi-layer and multi-chamber microchip system using thiol-ene ‘click chemistry’ aimed for drug transport studies across tissue barrier models. The fabrication process enables rapid prototyping of multi-layer microfluidic chips using different thiol-ene polymer mixtures, where porous Teflon membranes for cell monolayer growth were incorporated by masked sandwiching thiol-ene-based fluid layers. Electrodes for trans-epithelial electrical resistance (TEER) measurements were incorporated using low-melting soldering wires in combination with platinum wires, enabling parallel real-time monitoring of barrier integrity for the eight chambers. Additionally, the translucent porous Teflon membrane enabled optical monitoring of cell monolayers. The device was developed and tested with the Caco-2 intestinal model, and compared to the conventional Transwell system. Cell monolayer differentiation was assessed via in situ immunocyto-chemistry of tight junction and mucus proteins, P-glycoprotein 1 (P-gp) mediated efflux of Rhodamine 123, and brush border aminopeptidase activity. Monolayer tightness and relevance for drug delivery research was evaluated through permeability studies of mannitol, dextran and insulin, alone or in combination with the absorption enhancer tetradecylmaltoside (TDM). The thiol-ene-based microchip material and electrodes were highly compatible with cell growth. In fact, Caco-2 cells cultured in the device displayed differentiation, mucus production, directional transport and aminopeptidase activity within 9–10 days of cell culture, indicating robust barrier formation at a faster rate than in conventional Transwell models. The cell monolayer displayed high TEER and tightness towards hydrophilic compounds, whereas co-administration of an absorption enhancer elicited TEER-decrease and increased permeability similar to the Transwell cultures. The presented cell barrier microde-vice constitutes a relevant tissue barrier model, enabling transport studies of drugs and chemicals under real-time optical and functional monitoring in eight parallel chambers, thereby increasing the throughput compared to previously reported microdevices. © 2018 Tan et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
dc.rightsAttribution 4.0 International
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/
dc.sourceUnpaywall 20191101
dc.subjectaminopeptidase
dc.subjectdextran
dc.subjectinsulin
dc.subjectmannitol
dc.subjectmultidrug resistance protein 1
dc.subjectpolitef
dc.subjectrhodamine 123
dc.subjecttight junction protein
dc.subjectABC transporter subfamily B
dc.subjectABCB1 protein, human
dc.subjectdextran
dc.subjectinsulin
dc.subjectmaltose
dc.subjectmannitol
dc.subjectrhodamine 123
dc.subjecttetradecyl maltoside
dc.subjectArticle
dc.subjectbiocompatibility
dc.subjectbiological model
dc.subjectCaco-2 cell line
dc.subjectcell differentiation
dc.subjectcell growth
dc.subjectcontrolled study
dc.subjectdrug transport
dc.subjectelectric resistance
dc.subjectenzyme activity
dc.subjecthuman
dc.subjecthuman cell
dc.subjecthydrophilicity
dc.subjectimmunocytochemistry
dc.subjectintestine
dc.subjectmelting point
dc.subjectmicrofluidics
dc.subjectmonolayer culture
dc.subjectmucus
dc.subjectpermeability barrier
dc.subjectanalogs and derivatives
dc.subjectCaco-2 cell line
dc.subjectcytology
dc.subjectdevices
dc.subjectdrug effect
dc.subjectintestine absorption
dc.subjectintestine mucosa
dc.subjectlab on a chip
dc.subjectmetabolism
dc.subjectmicrofluidic analysis
dc.subjectprocedures
dc.subjectATP Binding Cassette Transporter, Sub-Family B
dc.subjectCaco-2 Cells
dc.subjectDextrans
dc.subjectHumans
dc.subjectInsulin
dc.subjectIntestinal Absorption
dc.subjectIntestinal Mucosa
dc.subjectLab-On-A-Chip Devices
dc.subjectMaltose
dc.subjectMannitol
dc.subjectMicrofluidic Analytical Techniques
dc.subjectRhodamine 123
dc.typeArticle
dc.contributor.departmentBIOMED INST FOR GLOBAL HEALTH RES & TECH
dc.description.doi10.1371/journal.pone.0197101
dc.description.sourcetitlePLoS ONE
dc.description.volume13
dc.description.issue5
dc.description.pagee0197101
Appears in Collections:Staff Publications
Elements

Show simple item record
Files in This Item:
File Description SizeFormatAccess SettingsVersion 
10_1371_journal_pone_0197101.pdf9.12 MBAdobe PDF

OPEN

NoneView/Download

Google ScholarTM

Check

Altmetric


This item is licensed under a Creative Commons License Creative Commons