A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies. / Tan, Hsih-Yin; Trier, Sofie; Rahbek, Ulrik L.; Dufva, Martin; Kutter, Jörg P.; Andresen, Thomas L.

In: PLoS ONE, Vol. 13, No. 5, e0197101, 2018.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Tan, H-Y, Trier, S, Rahbek, UL, Dufva, M, Kutter, JP & Andresen, TL 2018, 'A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies', PLoS ONE, vol. 13, no. 5, e0197101. https://doi.org/10.1371/journal.pone.0197101

APA

Tan, 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]. https://doi.org/10.1371/journal.pone.0197101

Vancouver

Tan H-Y, Trier S, Rahbek UL, Dufva M, Kutter JP, Andresen TL. A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies. PLoS ONE. 2018;13(5). e0197101. https://doi.org/10.1371/journal.pone.0197101

Author

Tan, Hsih-Yin ; Trier, Sofie ; Rahbek, Ulrik L. ; Dufva, Martin ; Kutter, Jörg P. ; Andresen, Thomas L. / A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies. In: PLoS ONE. 2018 ; Vol. 13, No. 5.

Bibtex

@article{adbfe32eed034a38812bc03a6a4bacc7,
title = "A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies",
abstract = "This paper presents the design and fabrication of a multi-layer and multi-chamber microchip system using thiol-ene {\textquoteleft}click chemistry{\textquoteright} 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.",
author = "Hsih-Yin Tan and Sofie Trier and Rahbek, {Ulrik L.} and Martin Dufva and Kutter, {J{\"o}rg P.} and Andresen, {Thomas L.}",
year = "2018",
doi = "10.1371/journal.pone.0197101",
language = "English",
volume = "13",
journal = "PLoS ONE",
issn = "1932-6203",
publisher = "Public Library of Science",
number = "5",

}

RIS

TY - JOUR

T1 - A multi-chamber microfluidic intestinal barrier model using Caco-2 cells for drug transport studies

AU - Tan, Hsih-Yin

AU - Trier, Sofie

AU - Rahbek, Ulrik L.

AU - Dufva, Martin

AU - Kutter, Jörg P.

AU - Andresen, Thomas L.

PY - 2018

Y1 - 2018

N2 - This 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.

AB - This 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.

U2 - 10.1371/journal.pone.0197101

DO - 10.1371/journal.pone.0197101

M3 - Journal article

C2 - 29746551

AN - SCOPUS:85046838279

VL - 13

JO - PLoS ONE

JF - PLoS ONE

SN - 1932-6203

IS - 5

M1 - e0197101

ER -

ID: 222244524