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

PLoS One. 2018 May 10;13(5):e0197101. doi: 10.1371/journal.pone.0197101. eCollection 2018.

Abstract

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 immunocytochemistry 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 microdevice 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.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • ATP Binding Cassette Transporter, Subfamily B / metabolism
  • Caco-2 Cells
  • Dextrans* / pharmacokinetics
  • Dextrans* / pharmacology
  • Humans
  • Insulin* / pharmacokinetics
  • Insulin* / pharmacology
  • Intestinal Absorption / drug effects*
  • Intestinal Mucosa / cytology
  • Intestinal Mucosa / metabolism*
  • Lab-On-A-Chip Devices*
  • Maltose / analogs & derivatives*
  • Maltose / pharmacokinetics
  • Maltose / pharmacology
  • Mannitol* / pharmacokinetics
  • Mannitol* / pharmacology
  • Microfluidic Analytical Techniques* / instrumentation
  • Microfluidic Analytical Techniques* / methods
  • Rhodamine 123* / pharmacokinetics
  • Rhodamine 123* / pharmacology

Substances

  • ABCB1 protein, human
  • ATP Binding Cassette Transporter, Subfamily B
  • Dextrans
  • Insulin
  • tetradecyl maltoside
  • Rhodamine 123
  • Mannitol
  • Maltose

Grants and funding

This work is supported by The Danish Council for Independent Research (FTP) and the Lundbeck Foundation; https://www.lundbeckfonden.com, https://dff.dk/en/front-page?set_language=en. None of the fund givers had any influence on the research.