Elsevier

Brain Research

Volume 853, Issue 1, 17 January 2000, Pages 115-124
Brain Research

Research report
Barrier function of porcine choroid plexus epithelial cells is modulated by cAMP-dependent pathways in vitro

https://doi.org/10.1016/S0006-8993(99)02317-3Get rights and content

Abstract

In the present paper, we demonstrate that the barrier properties of primary cultured epithelial cells isolated from porcine choroid plexus are regulated by cAMP-dependent signal transduction pathways in vitro. Triggering cAMP-connected cascades in cell layers grown on permeable filters with cAMP-analogues or forskolin led to a significant increase of transepithelial electrical resistances and a pronounced reduction in the permeation rate of a 4 kDa-dextran probe. In dose–response experiments using the cAMP-analogue 8-(4-chlorophenylthio)-cAMP transepithelial electrical resistances were observed to increase above a threshold concentration ranging between 10−5.5 and 10−5 M. Additional impedance studies performed with confluent cell layers grown on gold-film electrodes revealed that the observed changes in transepithelial resistances and presumably also in macromolecular permeation rates were not entirely caused by a reinforcement of intercellular junctions but also contained contributions from changes in the cell-substrate adhesion pattern. These inherent contributions to the electrical resistance and macromolecular permeability are caused by a restricted diffusion pathway between basal plasma membrane and culture substrate that have to be considered in data analysis, especially when leaky cell layers on filter substrates with low pore densities are used.

Introduction

Two cellular barriers effectively protect the mammalian brain from passive entrance of ions and hydrophilic compounds circulating in the bloodstream and thereby guarantee a constant chemical environment within the central nervous system (CNS). First, the capillaries of the brain are almost entirely lined by very unique endothelial cells that build up the so-called blood–brain barrier. Second, in very limited regions of the brain, where the endothelial cells do not provide a sufficiently tight barrier — namely in certain parts of the ventricular system, the choroid plexus — an underlying sheet of epithelial cells serves to separate blood from cerebrospinal fluid (blood–cerebrospinal-fluid barrier). The ability of both cell types to separate two compartments of different chemical composition arises from the formation of very tight intercellular junctions (tight junctions) [25]. These cell–cell contacts prevent diffusive permeation of blood derived compounds along the intercellular cleft between adjacent cells into the CNS or vice versa. Besides the sole barrier function, the choroid plexus epithelial cells, which are in the focus of the present study, are responsible for a multitude of physiological needs like active transport of micronutrients into the brain, clearance of certain toxic compounds out of the brain and eventually production of cerebrospinal fluid itself [5]. The choroid plexus is particularly important from a medical viewpoint since delivery of hydrophilic drugs to the CNS via the bloodstream is hampered by the same barriers that are responsible for CNS homeostasis as described above. Clearance activity of plexus cells and the expression of `multidrug-resistance' transport proteins [22] increase the difficulties to establish and maintain effective drug concentrations in the brain capable to treat neuronal diseases. Hence, there is considerable interest to understand the signal transduction pathways that control the barrier function of this epithelium in detail.

We recently introduced an in vitro model of the blood–cerebrospinal-fluid barrier based on choroid plexus epithelial cells isolated from porcine brain 9, 13, 14. When these cells are cultured on permeable filter inserts, the apical side of the cell layer mimics the ventricular compartment, whereas the basolateral side models the blood side. We were able to show that these cells maintain their characteristic structural and functional properties in vitro 9, 14. They express microvilli, ciliae and complex tight junctions and show active transport of micronutrients, antibiotics and fluid as well as the polar secretion of proteins that are unique for the cerebrospinal fluid [9]. In the present study, we explored the impact of the cAMP signal transduction cascade on the barrier function of these cells. The cAMP-cascade seemed challenging to us, as it is well known from epithelia and endothelia of other origins, that cAMP is often involved in barrier regulation 4, 8, 24, 28. It is furthermore known that the endothelial cells in cerebral capillaries, which fulfill similar physiological functions, respond to increased cAMP levels by barrier strengthening 6, 23.

Epithelial barrier function is commonly quantified by measuring the permeation rate of a marker compound of a certain molecular weight or by the very convenient determination of transepithelial electrical resistances (TER) as a measure of ionic permeability. Although both experimental approaches are well established and widely accepted, under some circumstances data interpretation may require some caution. Recently, Lo et al. [18] pointed out that the outcome of transepithelial resistance measurements for cell layers grown on porous filters may be affected by the separation distance between the basal cell membrane and the culture substrate. The reason for this is that any paracellular current that traverses the epithelium has to flow through the pores of the filter, then through the narrow channels between basal cell membrane and substrate, before it can escape through the paracellular shunt between adjacent cells. This current pathway underneath the cells provides an additional resistance that is strongly dependent on cell-substrate separation distance. It is inherently included in TER values determined by both the common DC-techniques or AC impedance analysis as long as cells are grown on a porated filter. Although not reported before, the same is true for permeability studies using macromolecular probes that have to diffuse along the same paracellular pathways as the ions in electrochemical experiments. However, when culturing the cells on top of solid gold-film electrodes, impedance analysis can distinguish between the resistance contribution arising from intercellular contacts between adjacent cells (barrier function) and the resistance that arises from cell-substrate adhesion [10]. This technique has been pioneered by Giaever and Keese 10, 11, and it is referred to as electric cell-substrate impedance sensing (ECIS).

In the present study, we analyzed the influence of elevated intracellular cAMP-levels on the barrier properties of choroid plexus epithelial cells grown on porous filters by measuring both transepithelial resistances and macromolecular permeabilities. Both methods consistently implicated a distinct increase of barrier efficiency. We additionally studied the response of plexus cells to cAMP-elevation when cells were grown on gold-film electrodes. Impedance analysis and subsequent data modeling confirmed that under these conditions, plexus cells strengthen their intercellular contacts and therefore enhance their barrier function. But we also observed significant changes associated with the cell-substrate contact zone that could not have been accounted for with the commonly used filter methods.

Section snippets

Cell preparation and culture conditions

Epithelial cells from porcine choroid plexus were prepared as already described elsewhere [9]. Briefly, choroid plexus tissue taken from freshly slaughtered pigs was incubated with 0.25% (w/v) trypsin solution (Biochrom, Berlin, Germany) for 2.5 h at 4°C and then for an additional 0.5 h at 37°C. Proteolytic activity was terminated by addition of newborn calf serum (Biochrom). Enzymatically unreleased tissue was separated from the cell suspension, and the released cells were spun down at 15×g.

Results

For the present study, we made use of an in vitro model of the blood–cerebrospinal fluid barrier (CSF) that is based on primary cultured choroid plexus epithelial cells derived from pig brain 9, 13, 14. Typical values for the cell layers' permeability towards macromolecular probes are 1.5·10−8 cm/s for 150 kDa-dextran and 5.0·10−7 cm/s for 4 kDa-dextran [14]. Transepithelial resistances were usually found between 100 and 150 Ω cm2. For the cell layers used here we determined average values

Discussion

The physiological regulation of epithelial and endothelial barrier function in the various organs of animal bodies has been the subject of numerous studies throughout the past years 3, 6, 8, 15, 24, 28. These investigations were mostly driven by the lack of understanding of how the cells comprising these interfacial tissues can adjust their barrier properties to particular physiological needs and also how pathological conditions manage to disrupt these anatomical barriers. Among others, the

Acknowledgements

This work has been financially supported by the German Ministry of Education and Research (BMBF) under contract No. 03114666B. The authors would like to thank Dr. I. Giaever and Dr. C.R. Keese (Rensselaer Polytechnic Institute, Troy, NY) for their support in impedance data modeling, careful revision of the manuscript and helpful discussions. A scholarship granted to J. Wegener by the Studienstiftung des Deutschen Volkes is gratefully acknowledged.

References (28)

  • H.F. Cserr

    Physiology of the choroid plexus

    Physiol. Rev.

    (1971)
  • M.A. Deli et al.

    Penetration of small molecular weight substances through cultured bovine brain capillary endothelial cell monolayers: the early effects of cyclic adenosine 3′,5′-monophosphate

    Exp. Physiol.

    (1995)
  • M.E. Duffey et al.

    Regulation of epithelial tight junction permeability by cAMP

    Nature

    (1981)
  • U. Gath et al.

    Porcine choroid plexus cells in culture: expression of a polarized phenotype, maintenance of barrier properties and apical secretion of CSF-components

    Eur. J. Cell Biol.

    (1997)
  • Cited by (68)

    • Independent impedimetric analysis of two cell populations co-cultured on opposite sides of a porous support

      2017, Experimental Cell Research
      Citation Excerpt :

      Impedance spectroscopy of cell-covered filter inserts has become a standard technique to investigate all kinds of barrier-forming cell types in the last decades [7,8]. Epithelial or endothelial cell layers are probed with respect to tight junctions formation during cell attachment and spreading [6] or changes in their barrier function in response to a certain stimulus [9–12]. The majority of these studies is performed with cell monolayers.

    • Label-free analysis of GPCR-stimulation: The critical impact of cell adhesion

      2016, Pharmacological Research
      Citation Excerpt :

      HEK293T-CRE-Luc-hH1R cells and HEK293T-CRE-Luc-hH1R-hMSR1 cells were studied by recording the impedance of the cell-covered electrodes along a frequency band from 10 Hz to 100 kHz (impedance spectroscopy). Frequency-resolved impedance data was analyzed according to a biophysical model that decomposes the overall impedance of a cell-covered electrode into subcellular contributions as described earlier [23,24]. According to this model, which is schematically illustrated in Fig. 2, the overall impedance of the cell layer arises from current flow along trans- (dashed lines) and paracellular ionic pathways (solid lines).

    • Real-time analysis of the effects of toxic, therapeutic and sub-therapeutic concentrations of digitoxin on lung cancer cells

      2014, Biosensors and Bioelectronics
      Citation Excerpt :

      Such studies do not only have to account for the narrow therapeutic window or drug׳s dose-dependent and selective apoptosis to cancer cells (Newman et al., 2008; Xie and Cai, 2003), but also need to allow monitoring of the cellular systems without lag time between sample collection and data analysis. Electric cell–substrate impedance sensing (ECIS) is a non-invasive and quantitative form of cell-based sensing that utilizes identical small gold-film electrodes deposited on the bottom of cell culture dishes to measure the cellular resistance and its alteration upon changes in cell morphology, spreading, attachment and migration (Arndt et al., 2004; Spegel et al., 2008), all as a function of applied frequencies and in real-time (Giaever and Keese, 1984; Wegener et al., 2000a). The applicability of ECIS was extended to inhibition assays for cytochalasin-D (cytoskeletal inhibitor) (Sapper et al., 2006), prostaglandin E2 (inflammatory mediator) (Wang et al., 1995), bacterial protease (Itagaki et al., 2011), or platelet-activating factors that affect in cellular adhesion (Melnikova et al., 2009).

    • Oral epithelial cell reaction after exposure to Invisalign plastic material

      2014, American Journal of Orthodontics and Dentofacial Orthopedics
      Citation Excerpt :

      This is a noninvasive technique that allows us to measure and record transepithelial resistance changes in real time. It is also sensitive enough to determine the cell-to-cell interactions such as barrier function and paracellular permeability.9,25 As previously mentioned, the electric cell-substrate impedance sensing system can report impedance values or translate impedance measurements to resistance measurements.

    • Label-free monitoring of cell-based assays: Combining impedance analysis with SPR for multiparametric cell profiling

      2013, Biosensors and Bioelectronics
      Citation Excerpt :

      For the more interested reader Fig. S3 (Supplementary material) provides an additional plot of the frequency-dependent total impedance of a cell-free electrode in comparison to a cell-covered electrode in complex presentation with the imaginary part of the impedance (reactance) plotted as a function of the real part (resistance). Taken together, these spectral characteristics are not significantly different from data recorded with commercial ECIS electrodes and they have been described by others before (Giaever and Keese, 1991, 1993; Wegener et al., 2000a). In complete agreement to the SPR measurements, the ECIS data confirm (i) the formation of a confluent MDCK II cell layer on the electrode surface and (ii) no perturbing influences of SPR detection on ECIS data recording.

    View all citing articles on Scopus
    1

    Present address: Department of Physics and Biology, School of Science, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA.

    2

    Present address: Department of Neurosurgery, Rhode Island Hospitals/Brown University, 593 Eddy Street, Providence, RI 02902, USA.

    View full text