You are on page 1of 9

..................................................................................................................................................................................................

Modulation of Cytokine Release by Differentiated


CACO-2 Cells in a Compartmentalized Coculture Model
with Mononuclear Leucocytes and Nonpathogenic
Bacteria
A. Parlesak*, D. Hallery, S. Brinz*, A. Baeuerlein* & C. Bode*

Abstract
*Department of Physiology of Nutrition, Institute To further investigate the interaction between human mononuclear leucocytes
for Biological Chemistry and Nutrition, [peripheral blood mononuclear cells (PBMC)] and enterocytes, the effect of a
Hohenheim University (140e), Stuttgart; yCentre confluent layer of differentiated CACO-2 cells on cytokine kinetics during
for Nutrition and Food Research, Immunobiology
of Nutrition, Technical University of Munich,
challenge with bacteria in a compartmentalized coculture model was investi-
Freising, Germany gated. Nonpathogenic Escherichia coli were added either to the apical or the
basolateral compartment of this transwell cell culture system, the latter of which
Received 23 February 2004; Accepted in revised contained human leucocytes. The synthesis of tumour necrosis factor (TNF-a)
form 17 June 2004 and interleukin (IL)-12 was significantly suppressed by CACO-2 cells when
Correspondence to: Alexandr Parlesak, PhD,
leucocytes were stimulated directly with bacteria. This suppression was not
Hohenheim University (140e), Department of paralleled by changes in the production of IL-10, IL-6 and transforming growth
Physiology of Nutrition, Garbenstraße 28, D-70593 factor (TGF)-b. When the bacteria were applied apically to the CACO-2 cell
Stuttgart, Germany. E-mail: parlesak@uni- layer, the production of TNF-a, IL-12, IL-1b, IL-8, IL-6, IL-10, TGF-b and
hohenheim.de interferon-g was pronouncedly lower as compared to the bacterial stimulation of
leucocytes beneath the CACO-2 cells. In the latter experiments, IL-6, IL-8 and
TNF-a were the cytokines being mostly induced by apical addition of E. coli.
Quantitative mRNA expression analysis revealed that IL-8 gene expression was
equally induced in both CACO-2 and PBMC after apical stimulation with
bacteria. Of note, bacteria-stimulated CACO-2 cells produced little or no
cytokines in the absence of leucocytes, supporting the concept of leucocyte–
epithelial cell cross-talk in modulating cytokine responses in the gut mucosa.

Introduction
molecules and their capability to present antigens by com-
Enteric bacteria are in close proximity to the intestinal ponents of the human major histocompatibility complex
epithelium as well as specialized, lymphoid and accessory [3–7]. We have previously shown that colonization of
cells in the lamina propria and the follicles of the gut- germ-free rats with Gram-negative, nonpathogenic, enteric
associated lymphoid tissue (GALT). The maintenance of bacteria leads to an early but transient activation of colonic
local homeostasis in the intestinal mucosa to enteric bac- epithelial cells [8, 9], demonstrating the physiological rele-
teria and their antigens requires a fine-tuned balance vance for commensal bacteria-induced signalling to the
between immune activation and regulation, a process mucosal epithelium.
that, if impaired, may lead to intestinal inflammation Increasing efforts are made on research dealing with
[1, 2]. Intestinal epithelial cells (IEC) are thought to con- effects of nutrients and drugs on the immune system
tribute to immunomodulation of mucosal leucocytes by at affecting the local immune response in the intestine. To
least two different mechanisms. Firstly, they separate bac- investigate the interaction among bacteria, enterocytes,
teria from intraepithelial lymphocytes and lamina propria and leucocytes, we developed a coculture model in which
mononuclear cells (LPMC). Secondly, IEC can trigger the a confluent layer of differentiated enterocytes (CACO-2
immune response in GALT by the secretion of cytokines, cells) separates an apical compartment with nonpathogenic
the constitutive or inducible expression of costimulatory bacteria from a basolateral compartment with

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485 477
478 Coculture of Bacteria, Enterocytes and PBMC A. Parlesak et al.
..................................................................................................................................................................................................

mononuclear leucocytes that were isolated from human Millicell ERS Ohmmeter, Millipore, Eschborn, Germany)
peripheral venous blood [peripheral blood mononuclear and visual control of cell layer integrity under the micro-
cells (PBMC)] [10]. In this model, leucocyte–epithelial scope. Cells became confluent after 6  1 days. Within this
cell cross-talk became evident [8–11]. time, the TEER exceeded the cut-off point of 450 O  cm2
The enterocyte-like cell line CACO-2 used in the pre- [10]. After complete differentiation, which lasted for further
sent model mimics important properties of the mucosal 10  2 days, coincubation experiments were performed.
epithelium; after confluence, cells stop to proliferate and Bacteria culture. Nonpathogenic Escherichia coli K12
spontaneous polarized differentiation occurs [12]. After were precultured (37  C) on Luria broth (1% Trypton:
differentiation, microvilli, desmosomes and tight junctions Oxoid, Wesel, Germany; 0.5% yeast extract: Difco,
are formed [13], and enzymes of the brush border mem- Heidelberg, Germany; 0.5% NaCl) agar plates. Thereafter,
brane are expressed [14]. The permeability of a CACO-2 2  10 ml of liquid Luria broth was inoculated from the
cell monolayer to macromolecules [13] and bacterial pro- agar plates. After an additional 12 h, 100 ml of the incubation
ducts such as endotoxin [10] was shown to be absent or medium were transferred into fresh Luria broth medium
negligible after confluence and differentiation. and incubated further for 24 h to bring the bacteria into
In the present study, we tested the hypothesis whether the stationary growth phase. Thereafter, the bacteria sus-
or not the presence of IEC affects the cytokine production pension was centrifuged and bacteria were washed twice
of human leucocytes and, if so, to which extent this with phosphate-buffered saline. To obtain the final bac-
immunomodulation occurs. Furthermore, the role of spa- teria stock solution, cell concentration was adjusted to
tial separation of bacteria and leucocytes by the IEC layer 5.0  109 colony-forming units (cfu)/ml in cell culture
was investigated. For this purpose, the kinetics of 11 medium. Gentamicin (120 mg/ml; Gibco BRL/Life Tech-
different cytokine concentrations were monitored for nologies) was added to the medium to avoid uncontrolled
64 h (in some experiments up to 72 h) in the basal com- growth [10].
partment of the described model. Out of these cytokines, Leucocyte preparation. Mononuclear leucocytes from
three can be contributed to acute inflammation [tumour peripheral venous blood (PBMC) of healthy donors (two
necrosis factor (TNF)-a, interleukin (IL)-1b and IL-8], women, eight men; age: 42  10 years) were isolated from
three to activation of TH1-type lymphocytes [IL-12, inter- buffy coats (cell-enriched plasma) that were obtained from
feron (IFN)-g, IL-2] and two to activation of TH2-type the Institute of Blood Transfusion Medicine, Katharinen-
lymphocytes (IL-4, IL-5) [15]. Furthermore, the kinetics hospital Stuttgart. Equal volumes (15 ml) of cell-enriched
of three cytokines [IL-6, IL-10 and transforming growth plasma and RPMI 1640 medium were mixed and layered
factor (TGF)-b1] was measured, which can act either over 15 ml Ficoll solution (r ¼ 1.077 g/ml; both solutions:
stimulating or suppressive on immunocompetent cells, Seromed, Berlin, Germany). After centrifugation (400  g,
depending on the kind and state of cell being affected. 30 min), mononuclear leucocytes were taken from the
interface, washed three times with RPMI 1640 medium
and adjusted to 2.0  107 cells/ml.
Materials and methods
Cocultivation and stimulation with bacteria. To investi-
Cell culture. The human enterocyte-like cell line CACO-2 gate the interaction among bacteria, differentiated CACO-
was obtained from the ‘Deutsche Sammlung von Mikroor- 2 cells and human leucocytes, six different experimental
ganismen und Zellkulturen’ (Braunschweig, Germany: approaches were conducted. Each experiment was repeated
DSMZ number ACC169) and was cultured in Dulbecco’s 10 times in triplicate with PBMC (4.0  106) from 10
Modified Eagle Medium (DMEM) containing 17% inac- healthy donors. In two further experiments, the differen-
tivated (56  C for 30 min) fetal calf serum (FCS) and 1% tiated CACO-2 cell layer was incubated either with or
concentrated nonessential amino acid solution (all without bacteria in the absence of leucocytes. Total
components: Gibco BRL/Life Technologies, Karlsruhe, volume in the basal and apical compartment was always
Germany). FCS was controlled for endotoxin content 2.0 ml. Bacterial stimulation was performed with E. coli
(<0.2 ng/ml: LAL Test, Chromogenix, Mölndal, Sweden). K12 (2.0  107 cfu per compartment). In detail, experi-
For transwell cultures, 1.0  106 cells were suspended in ments were as follows (Fig. 1):
2 ml cell culture medium and cultured in inserts on a
semipermeable polyethylene terephthalate membrane 1. To obtain the maximum stimulation of the applied leuco-
(4.3 cm2 surface, 0.45 mm pore diameter, 1.6  106 cytes under comparable conditions, PBMC were stimu-
pores/cm2: Falcon, Becton-Dickinson, Le Pont De Claix, lated by direct contact with E. coli K12 (no CACO-2 cells
France). Inserts were placed into cavities of six-well plates were present).
(Greiner, Frickenhausen, Germany). During cell growth 2. To assess the effect of CACO-2 cells on bacteria-induced
and differentiation, medium in both compartments was secretion of cytokines by leucocytes, the E. coli
changed three times a week. Confluence was controlled by K12–PBMC suspension was coincubated beneath inserts
measurement of transepithelial electrical resistance (TEER; with confluent CACO-2 cells.

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485


A. Parlesak et al. Coculture of Bacteria, Enterocytes and PBMC 479
..................................................................................................................................................................................................

A B

C D

Figure 1 Experimental arrangements in the transwell system for the assessment of leucocyte–enterocyte interactions under different kinds of stimulation
by nonpathogenic Escherichia coli. Leucocytes were challenged with bacteria either in the absence of CACO-2 cells (A) or beneath a differentiated
monolayer of these cells (B). CACO-2 cells were stimulated apically with E. coli either in the presence of PBMC (C) or without leucocytes (D).

3. Bacteria in the apical compartment were separated from Permeability of CACO-2 cell monolayers. To assess the
PBMC in the basolateral compartment by a confluent integrity of the confluent CACO-2 cell layer, fluorescein-
CACO-2 monolayer. labelled dextran Mr 4400 (Sigma-Aldrich, Steinheim,
4. To assess the basal secretion of cytokines, PBMC were Germany) was added to the apical compartment in
cultured beneath the differentiated CACO-2 cells on inserts experimental approaches no. 3, 4, 5 and 6. Relative perme-
in the absence of bacteria (negative control experiments). ability of this marker was measured in the basal medium
5. The contribution of the CACO-2 cells to the production with a microplate fluorescence reader (BMG, Freiburg,
of cytokines was assessed by stimulating differentiated Germany).
CACO-2 cells on inserts with bacteria as described above RNA isolation and Light Cycler analysis. RNA was isolated
in the absence of leucocytes. using Trizol (Invitrogen, Karlsruhe, Germany) and 1 mg of
6. The basal production of cytokines by CACO-2 cells (with- total RNA was reverse transcribed as described previously
out bacteria and leucocytes) and, separately, by PBMC [9]. Quantitative polymerase chain reaction (PCR) was done
(without CACO-2 cells and bacteria) was monitored as in the Light CyclerTM using LC-FastStart DNA Master
well. KitTM (Roche, Penzberg, Germany) and the following pri-
mers: IL-8-A; (50 ) 5-ATGACTTCCAAGCTGGC-3 and
The incubations with PBMC were aborted after 4 h, 8 h, IL-8-B; (30 ) 5-ACTTCTCCACAACCCT-3, 18SrRNA-A;
16 h, 24 h, 32 h and 64 h, those without leucocytes after (50 ) 5-AAGTCTTTGGGTTCCGGG-3 and 18SrRNA-B
12 h, 24 h and 72 h. For the estimation of the reduction of (30 ) 5-GGACATCTAAGGGCATCACA-3. PCR amplifi-
cytokine production by sole spatial separation by the semi- cation produced a 274 and 205 bp product, respectively.
permeable membrane, bacteria were added to the apical The PCR program was one cycle of denaturation at
side of the inserts (without CACO-2 cells) which were 95  C for 10 min followed by 50 cycles of 95  C for 15 s,
placed above the PBMC suspension. This experiment annealing at 60  C for 10 s and extension at 72  C for 20 s.
was abrogated after 16 h. The amplified product was detected by the presence of a

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485


480 Coculture of Bacteria, Enterocytes and PBMC A. Parlesak et al.
..................................................................................................................................................................................................

SYBR1 green fluorescent signal. Melting curve analysis and IL-6 and IL-8 increased continuously after bacterial
gel electrophoresis were used to document the amplicon stimulation, the latter having a biphasic course
specificity. Calibration curves were generated by measuring (Fig. 2). The onset of the production of the TH1-type
serial dilutions of stock cDNA. For the relative quantifica- cytokines IFN-g and IL-2 was delayed compared to the
tion of IL-8 gene expression in CACO-2 and PBMC, the other cytokines. In all experiments, IL-8 and IL-6 were
crossing point (Cp) of the log-linear portion of the ampli- the dominant mediators reaching by far the highest
fication curve was determined. IL-8/18S ribosomal RNA concentrations out of the measured cytokines (Fig. 2,
ratios were used to normalize the data. The data are pre- Table 1). Production of cytokines by nonstimulated
sented as fold increase of IL-8 mRNA in relation to the PBMC in the absence of CACO-2 cells was either not
nonstimulated controls including CACO-2 and PBMC, detectable [TGF-b (32 h); IL-12 (32 h)] or very low
respectively. [TNF-a (12 h): 7.3  3.9 pg/ml; IL-10 (32 h):
Measurement of cytokine concentrations. In all experi- 5.2  1.0 pg/ml; IL-8 (72 h): 4.48  1.03 ng/ml; IFN-g
ments with compartmentalization, cytokines were meas- (72 h): 23.8  3.3 pg/ml; IL-2 (72 h): 13.4  3.4 pg/
ured in the basal medium, because in previous ml)]. IL-4 and IL-5 were not detectable in any of the
experiments, relevant cytokine concentrations occurred samples mentioned above.
only in the basolateral compartment [10]. After centrifu-
gation of the medium (2000  g, 2 min), the supernatant
Bacterial stimulation of PBMC in the presence of CACO-2 cells
was frozen in aliquots (80  C). The following cytokines
were measured: TNF-a, IL-1b, IL-2, IL-4, IL-5, IL-6, The addition of differentiated CACO-2 cells on transwell
IL-8, IL-10, IL-12, IFN-g and TGF-b1. Out of the cyto- inserts above the PBMC–E.coli suspension resulted in a
kines measured, TGF-b1 was the only one being already significantly reduced production of TNF-a and IL-12
present in the FCS used for incubation of cells. Therefore, (Fig. 2). Moreover, IL-1b synthesis was transiently sup-
only the increase in concentration of this cytokine was pressed early after bacterial stimulation (12 h). The differ-
regarded in the evaluation. All cytokine concentrations entiated CACO-2 cell layer also induced a shift in the
were measured with OptEIA ELISA sets (BD Pharmingen, biphasic IL-8 secretion course, demonstrating a delayed
Heidelberg, Germany) according to the instruction of the onset of IL-8 production in bacteria-stimulated PBMC
manufacturer. (Fig. 2). In contrast, the kinetics of IL-10, IL-6, TGF-b,
Statistics. If not indicated otherwise, values are given IFN-g and IL-2 were not significantly affected by the
as means  standard error of the mean (SEM). Before CACO-2 cells, suggesting differential effects of the epithe-
evaluation of cytokine profiles with ANOVA (within-sub- lial cell monolayer on bacteria-induced cytokine secretion
ject design, sixfold-repeated measurements), values patterns in PBMC.
were transformed logarithmically to obtain normal dis-
tribution and homogeneity of variances. Experiments
Apical stimulation of CACO-2 cells by bacteria in the
without kinetic measurements were evaluated with a
basolateral presence of PBMC
one-way ANOVA and Tukey’s post hoc test. Only if
statistical difference (P < 0.05) between cytokine con- In experiments where leucocytes and bacteria were separ-
centrations occurred, they were termed as ‘higher’ or ated by the semipermeable membrane and the confluent
‘lower’. CACO-2 cell layer, the final concentration of all cytokines
mentioned above (besides IL-2) was significantly reduced
as compared to experiments where leucocytes were stimu-
lated by direct contact with bacteria (Fig. 2). Remarkable
reductions were achieved for the production of TNF-a
Results
(8 h: 98%), IL-1b (24 h: 98%), IL-6 (64 h: 96%),
IL-10 (32 h: 97%), IFN-g (64 h: 95%), IL-12 (24 h:
Bacterial stimulation of PBMC
89%) and IL-8 (24 h: 82%). The smallest difference
Four different types of cytokine secretion kinetics were was obtained for TGF-b (32 h:  45%; Fig. 2). Interest-
distinguishable in bacteria-stimulated leucocyte cocul- ingly and in contrast to all cytokines mentioned above, the
tures. TNF-a was the only cytokine that reached max- production of IL-2 after 64 h was significantly higher as
imal concentration between 8 h and 16 h, and the compared to experiments where leucocytes had direct con-
concentration of which decreased from that time stea- tact with bacteria (64 h: þ600%) (Fig. 2).
dily. IL-1b, IL-10, TGF-b and IL-12 secretion Of note, in experiments with bacterial challenge of
increased continuously after initial stimulation. Maxi- enterocytes, the concentrations of TNF-a, IL-1b, IL-8,
mal concentrations of these cytokines were reached after IFN-g, IL-10 and IL-6 were significantly higher as com-
24–32 h of stimulation and remained relatively constant pared to experiments without bacteria (Table 1). Cytokines
until the end of the incubation. The concentration of being especially induced by this challenge were IL-6 (32 h:

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485


A. Parlesak et al. Coculture of Bacteria, Enterocytes and PBMC 481
..................................................................................................................................................................................................

6 400

20
5
300
15 4

IL-12 (pg/ml)
TNF-α (ng/ml)

IL-10 (ng/ml)
3 200
10

2
100
5
1

0 0 0
0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70

Time (h) Time (h) Time (h)

15 200 5

12 160 4
IL-1β (ng/ml)

IFN-γ (ng/ml)
IL-6 (ng/ml)

9 120 3

6 80 2

3 40 1

0 0 0
0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70

Time (h) Time (h) Time (h)


1600
1400 180
Newly synthesized TGF-β1 (pg/ml)

1200 150
1200
1000
120
IL-2 (pg/ml)
IL-8 (ng/ml)

800
800 90
600
60
400 400

200 30

0 0 0
0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70

Time (h) Time (h) Time (h)

Figure 2 Concentrations of nine different cytokines [tumour necrosis factor-a, interleukin (IL)-1b, IL-8, IL-10, IL-6, transforming growth factor-b1,
IL-12, interferon-g and IL-2] within the first 64 h of cocultivation of enterocytes, bacteria (Escherichia coli K12) and mononuclear leucocytes [peripheral
blood mononuclear cells (PBMC)]. Bacteria were added either into the apical (&, dashed line) or the basolateral (*, solid line) compartment with respect
to the differentiated CACO-2 cell monolayer. In the third experimental arrangement (*, dotted line), bacteria and PBMC had direct contact and the
CACO-2 cell monolayer was omitted.

about 880-fold), TNF-a (8 h: about 51-fold) and IL-8 both CACO-2 cells (relative fold increase 15.4) and
(24 h: 36-fold). Cytokines being only moderately induced PBMC (relative fold increase 10.4) after 12 h of stimula-
by apical stimulation in the compartmentalized model tion. Nevertheless, Cp values significantly differed in
were IL-12, IL-2 and TGF-b (Table 1). CACO-2 cells (Cp ¼ 24.0) and PBMC (Cp ¼ 18.3),
To investigate the cellular source of cytokines, we cocul- suggesting that the absolute quantity of IL-8 mRNA is
tured CACO-2 with PBMC from five different donors much higher in PBMC. After 4 h of bacterial stimulation,
and stimulated the CACO-2 cell monolayer with E. coli IL-8 mRNA expression was only induced in PBMC (rela-
K12 for 4 and 12 h. Quantitative analysis of IL-8 mRNA tive fold increase 3.1) but not in CACO-2 cells (relative
expression in CACO-2 cells and PBMC was performed by fold increase 0.9).
using Light Cycler analysis. As shown in Fig. 3, IL-8 In control experiments where bacteria and PBMC were
mRNA expression was comparably strongly induced in separated by the semipermeable membrane only (without

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485


482 Coculture of Bacteria, Enterocytes and PBMC A. Parlesak et al.
..................................................................................................................................................................................................

Table 1 Time course of the concentration of nine cytokines in the basolateral compartment of the coculture model in the presence of PBMC from 10
donors with and without Escherichia coli K12 used as an apical challenge of the CACO-2 cell monolayer

Cytokine Challenge (yes/no) 4h 8h 16 h 24 h 32 h 64 h


TNF-a (pg/ml) Control 31 53 18  8 12  3 82 51
E. coli K12 123  27 257  32 221  35 76  14 27  7 92
IL-1b (pg/ml) Control 80  17 78  12 51  14 87  23 70  7 66  8
E. coli K12 62  22 63  19 230  78 206  62 259  59 158  58
IL-8 (ng/ml) Control 0.66  0.23 1.37  0.30 8.79  4.50 3.26  0.85 3.14  0.94 4.56  0.99
E. coli K12 3.20  0.80 9.23  1.73 50.1  10.8 118  27.7 86.0  13.5 157  18.7
IL-12 (pg/ml) Control ND ND ND ND ND ND
E. coli K12 15  4 93 12  4 19  4 12  3 15  5
IFN-g (pg/ml) Control 73 73 82 10  1 13  3 16  4
E. coli K12 51 51 12  3 20  5 31  6 156  33
IL-2 (pg/ml) Control 20  4 22  5 32  3 41  9 46  10 98  24
E. coli K12 41 72 27  5 42  6 50  8 147  25
IL-10 (pg/ml) Control 31 82 23  13 16  5 72 14  4
E. coli K12 61 27  4 119  19 164  25 139  20 129  21
IL-6 (pg/ml) Control 25  12 16  4 11  8 81  53 12  5 22  6
E. coli K12 380  103 2240  520 8210  1880 11640  2950 10520  2220 9320  2260
TGF-b1 (pg/ml) Control 171  53 181  64 278  62 317  81 479  107 342  114
E. coli K12 237  44 254  45 329  49 520  88 582  97 600  119

TNF, tumour necrosis factor; IL, interleukin; TGF, transforming growth factor; ND, not detectable.

CACO-2 cells), the synthesis of TNF-a and IL-10 were separated by the confluent CACO-2 cell layer
(6.64  1.06 ng/ml and 1.58  0.20 ng/ml, respectively) (Table 1).
after 16 h was about 38 and 65% compared to that pro-
duced by suspensions of leucocytes and bacteria without
CACO-2 cells, but still 30 and 13 times higher, respect- Bacterial stimulation of CACO-2 cells in the absence of PBMC
ively, compared to incubations where PBMC and bacteria In the absence of leucocytes, the nonchallenged CACO-2
cell layer secreted negligible amounts of TNF-a, IL-10,
IL-6 and IL-8 into the basal compartment (Fig. 4). The
22 concentration of all these cytokines increased significantly
20 after apical stimulation of the enterocyte-like cells with
18 bacteria (Fig. 4), but the concentrations remained far
16 below those occurring after bacterial challenge of the apical
(Fold increase)

14 surface of the CACO-2 cell layer in the basolateral pre-


IL-8 mRNA

12 sence of leucocytes (Table 1). After 72 h, IL-2 and IFN-g


10 were below detection limit in incubations without leuco-
8 cytes in the basolateral compartment.
6
4
2 Transepithelial electrical resistance/permeability to dextran Mr
0 4400
4 12 4 12 Time (h)
TEER of the CACO-2 cell monolayer increased during
CACO-2 PBMC development of confluence and differentiation by
641  85 O  cm2. The lowest increase (522 O  cm2)
Figure 3 Quantitative mRNA expression analysis for interleukin (IL)-8 in
CACO-2 cells and peripheral blood mononuclear cells (PBMC). CACO- clearly exceeded the suggested cut point of 450 O  cm2
2 cells were stimulated with Escherichia coli K12 for 4 h and 12 h in the [10]. The microscopic investigation of the basolateral
presence of PBMC in the basolateral compartment of the transwell cell medium revealed that neither bacteria nor CACO-2 cells
culture system. RNA was isolated and reverse transcribed as described in were able to migrate through the PET membrane. The
Material and methods. IL-8 mRNA expression was quantified using Light
integrity of the CACO-2 cell monolayer remained fairly
Cycler analysis and presented as fold increase relative to nonstimulated
control cells. The data in the bar chart represent the mean  SEM from stable for at least 24 h, as assessed by its permeability to
five different experiments with PBMC from five different healthy dextran Mr 4400 (Fig. 5). Surprisingly, the permeability of
volunteers. the CACO-2 cell layer increased significantly after 72 h

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485


A. Parlesak et al. Coculture of Bacteria, Enterocytes and PBMC 483
..................................................................................................................................................................................................

300 8

250 *

Permeability of dextran (%)


6
Concentration (pg/ml)

200

4
150

100 2

50
0
0 20 40 60 80
0
Time (h)
TNF (12 h) IL-10 (24 h) IL-6 (24 h) IL-8 (24 h)
Figure 5 Relative permeability of fluorescein isothiocyanate-labelled
Figure 4 Secretion of tumour necrosis factor-a, interleukin (IL)-10, IL-6
dextran Mr 4400 through the CACO-2 cell monolayer over 72 h. The
and IL-8 by the CACO-2 layer without and with bacteria on the apical
permeability was measured in the CACO-2 cell layer without other cells
surface after 12 h/24 h in experiments without leucocytes (*P < 0.05).
(*, dotted line), in the CACO-2 cell layer with peripheral blood
mononuclear cells (PBMC) in the basal compartment (&, dashed line), in
the CACO-2 cell layer with bacteria on the apical side (, dashes and
only if both bacteria and PBMC were present, but not
dots) and in the CACO-2 cell layer with bacteria on the apical side and
after the addition of bacteria alone. PBMC in the basal medium (&, solid line) (*P < 0.05).

Discussion
stimulation of PBMC in our model system may reflect
We have previously shown that nonpathogenic commensal more pathological conditions. Consequently, with the
enteric bacteria have the potential to initiate innate host exception of IL-2, IL-4 and IL-5, the separation of bacteria
responses in IEC in vivo and in vitro [8, 9]. To mimic the and PBMC by the confluent layer of differentiated
complex interaction of the epithelium with adjacent CACO-2 cells resulted in a pronounced reduction of
immune cells in the intestinal mucosa, we established a cytokine secretion into the basolateral compartment
reductionist epithelial–immune cell coculture system, (Fig. 2). Only a small part of this effect can be contributed
demonstrating a role for PBMC in initiating and regulat- to sole spatial separation by the semipermeable PET mem-
ing bacteria-mediated activation of IEC [8–11]. Previous brane, which reduced the production of TNF-a and IL-10
studies showed that IEC profoundly inhibit monocyte by the factor of 2-3 (after 16 h). Owing to the specifica-
activation [11, 16, 17] and T-cell function [18] through tions of the manufacturer, the semipermeable PET mem-
both secreted mediators and cell–cell interactions. In the brane has pores with a diameter <0.4 mm, which does not
present study, we show immunosuppressive effects of allow the permeation of intact bacteria through the mem-
CACO-2 cells on E. coli-stimulated PBMC. The secretion brane, but enables the diffusion of bacterial fragments and
of the pro-inflammatory cytokines TNF-a, IL-12 and, to products. This diffusion seems to be strongly reduced by
some extent, IL-1b was significantly inhibited. On the the CACO-2 cell monolayer, as the permeation of mol-
other hand, cytokines such as IL-10 and TGF-b1, which ecules with a molecular mass of Mr 4400 is clearly limited,
can suppress the synthesis of pro-inflammatory cytokines at least within the first 24 h of incubation (Fig. 5). There-
[19–21], were not significantly affected by the presence of fore, the suppression of diffusion of macromolecular pro-
CACO-2 cells. Direct contact between CACO-2 cells and ducts from the apical compartment is likely to contribute
PBMC was prevented by the use of 0.4 mm pore size substantially to the reduction of cytokine production
membranes in transwell inserts, suggesting that immuno- resulting from the separation of bacteria and leucocytes
suppressive factors were secreted from CACO-2 cells. by the CACO-2 cell monolayer. The combined barrier of
However, from the present experiments, the factor respon- membrane and CACO-2 cells reduced the synthesis of
sible for the suppression of TNF-a, IL-12 and IL-1b these cytokines by factors up to 82 (TNF-a), supporting
synthesis remains unknown. the concept that the intestinal epithelium contributes to
Translocated bacteria and their products are thought to maintain an immunosuppressive environment in the nor-
participate in initiating and perpetuating chronic mucosal mal gut [25]. Surprisingly, neither the apical addition of
inflammation [22–24], suggesting that the direct bacterial bacteria alone nor the basal addition of PBMC alone

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485


484 Coculture of Bacteria, Enterocytes and PBMC A. Parlesak et al.
..................................................................................................................................................................................................

seemed to affect the integrity of the CACO-2 cell mono- marker through the layer of CACO-2 cells, a transport
layer within the applied incubation time (Fig. 5). The of intact bacterial antigens through this barrier seems
increase in permeability in the presence of both bacteria unlikely. Hence, the mechanism for the enhanced IL-2
and PBMC after 72 h was paralleled by an increase of the production by PBMC in the presence of CACO-2
concentration of IL-8, TNF-a and IFN-g. In previous cells remains unknown. Noteworthy in this context are
studies, these cytokines were shown to impair epithelial the occurrence of intestinal inflammation in mice with a
integrity [26–29], which makes them possible mediators of disrupted IL-2 gene [37] and the promotion of intestinal
the increased permeability measured in the present study. cell restitution by IL-2 [38]. The current opinion that
We have previously shown that Gram-negative bacteria activation of TH1 cell results in the parallel shedding of
induce nuclear factor (NF)-kB signalling and kB-depend- characteristic cytokines (IL-12, IL-2 and IFN-g) [15]
ent gene expression in IEC. Considering that the stimula- seems not to be met within the presented model, as IL-2
tion of CACO-2 cells with bacteria in the presence of on the one hand and IFN-g together with IL-12 on the
PBMC still induced significant IL-8 and IL-6 protein other are differently regulated.
production and that both genes are regulated at least in Evidently, the applied strain of E. coli can be considered as
part by the transcription factor NF-kB [30] and are a standard stimulus only and cannot meet the complex
expressed in IEC [9, 10], we next evaluated the cellular properties of the intestinal microflora. Therefore, long-term
source of cytokine production by performing quantitative interactions between intestinal flora and the epithelium of
mRNA expression analysis in CACO-2 and PBMC. Inter- the intestine, which are also affected by bacterial metabolism,
estingly, IL-8 gene expression was induced in CACO-2 cannot be investigated in the described model, mainly
and PBMC at an early stage of cocultivation (12 h), the because the intestinal flora cannot be cultivated outside the
total amount of IL-8 mRNA being distinctly higher in gut for obvious reasons. Instead, in the present model, a
PBMC. Taken together, the complex nature of leucocyte– standardized, short-termed immunological stimulation
epithelial cell cross-talk makes it difficult to speculate occurs, which regards leucocyte–enterocyte interactions, the
about the absolute contribution of each cell type in the modulation of which by drugs and nutrients can be
production of IL-8 protein in the coculture model. Con- measured.
versely, CACO-2 cells are not capable of IFN-g and IL-2
production within the applied model [10], and the
presence of PBMC did not induce synthesis of the latter Acknowledgments
cytokines in IEC (data not shown). We thank Dr Luz from the Institute of Blood Transfusion
Enterocyte-like cell lines were used as models to obtain Medicine, Katharinenhospital Stuttgart, who made the
important information about the involvement of IEC in buffy coats available. We are indebted to Daniela Distel
the immune response of the intestinal mucosa, especially for culturing cells and bacteria and to Susanne Scharr and
to evaluate the pathology of enteropathogens [4, 31, 32]. It Sabine Fischer for their support in the measurement of
is our understanding that epithelial–immune cell interac- cytokines. This work was supported by the Wissenschafts-
tions contribute to the homeostasis of normal intestinal förderung der Deutschen Brauwirtschaft e. V. (Project
mucosa [25]. In the present model, although some tissue- B67) and by the young investigator award for Dirk Haller
specific cells present in the GALT such as lamina propria in the Emmy Noether program by the Deutsche For-
lymphocytes, intraepithelial lymphocytes, mast cells, den- schungsgemeinschaft (HA 3148/1-2).
dritic cells and other cells are missing, the model allows the
assessment of leucocyte–enterocyte interaction. Previous
studies showed that freshly isolated nonstimulated LPMC References
secrete IL-6 (approximately 5 ng/106 cells/48 h of incuba-
1 Blumberg RS, Saubermann LJ, Strober W. Animal models of muco-
tion) [33], TNF-a (215 pg/106 cells/24 h) [34] and IL-8 sal inflammation and their relation to human inflammatory bowel
(approximately 4.5 ng/106 cells/24 h) [35] in similar quan- disease. Curr Opin Immunol 1999;11:648–56.
tities compared to the concentrations measured in the 2 Sartor RB. Pathogenesis and immune mechanisms of chronic
basolateral compartment of CACO-2/PBMC cocultures inflammatory bowel diseases. Am J Gastroenterol 1997;92 (12
after apical stimulation with bacteria (Table 1). Our recent Suppl.):5S–11S.
3 Sartor RB. Cytokines in intestinal inflammation: pathophysiological
data also indicate similar function of LPMC and PBMC in and clinical considerations. Gastroenterology 1994;106:533–9.
the regulation of epithelial cell responses to bacterial sti- 4 Kagnoff MF, Eckmann L. Epithelial cells as sensors for microbial
muli ex vivo in the coculture system [36]. However, the infection. J Clin Invest 1997;100:6–10.
processes within the GALT leading to specific immunity 5 Blumberg RS, Terhorst C, Bleicher P et al. Expression of a
cannot be mimicked by the present model. This holds true nonpolymorphic MHC class I-like molecule, CD1D, by human
intestinal epithelial cells. J Immunol 1991;147:2518–24.
especially for the production of IL-2 which is generally 6 Panja A, Barone A, Mayer L. Stimulation of lamina propria lym-
considered to require specific T-cell receptor activation. phocytes by intestinal epithelial cells: evidence for recognition of
According to the negligible permeation of the permeability nonclassical restriction elements. J Exp Med 1994;179:943–50.

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485


A. Parlesak et al. Coculture of Bacteria, Enterocytes and PBMC 485
..................................................................................................................................................................................................

7 Hershberg RM, Framson PE, Cho DH et al. Intestinal epithelial 22 Sedman PC, Macfie J, Sagar P et al. The prevalence of gut transloca-
cells use two distinct pathways for HLA class II antigen processing. J tion in humans. Gastroenterology 1994;107:643–9.
Clin Invest 1997;100:204–15. 23 Duchmann R, May E, Heike M, Knolle P, Neurath M, Meyer
8 Haller D, Russo MP, Sartor RB, Jobin C. IKK beta and phospha- zum Buschenfelde KH. T cell specificity and cross reactivity
tidylinositol 3-kinase/Akt participate in non-pathogenic Gram-nega- towards enterobacteria, bacteroides, bifidobacterium, and antigens
tive enteric bacteria-induced RelA phosphorylation and NF-kappa B from resident intestinal flora in humans. Gut 1999;44:812–8.
activation in both primary and intestinal epithelial cell lines. J Biol 24 Sartor RB. The influence of normal microbial flora on the develop-
Chem 2002;277:38168–78. ment of chronic mucosal inflammation. Res Immunol
9 Haller D, Holt L, Kim SC, Schwabe RF, Sartor RB, Jobin C. 1997;148:567–76.
Transforming growth factor-beta 1 inhibits non-pathogenic Gram 25 Strober W. Interactions between epithelial cells and immune cells in
negative bacteria-induced NF-kappa B recruitment to the interleu- the intestine. Ann N Y Acad Sci 1998;859:37–45.
kin-6 gene promoter in intestinal epithelial cells through modulation 26 Sansonetti PJ, Arondel J, Huerre M, Harada A, Matsushima K.
of histone acetylation. J Biol Chem 2003;278:23851–60. Interleukin-8 controls bacterial transepithelial translocation at the
10 Haller D, Bode C, Hammes WP, Pfeifer AM, Schiffrin EJ, Blum S. cost of epithelial destruction in experimental shigellosis. Infect
Non-pathogenic bacteria elicit a differential cytokine response by Immun 1999;67:1471–80.
intestinal epithelial cell/leucocyte co-cultures. Gut 2000;47:79–87. 27 Seydel KB, Li E, Zhang Z, Stanley SL Jr. Epithelial cell-initiated
11 Haller D, Serrant P, Peruisseau G et al. IL-10 producing CD14low inflammation plays a crucial role in early tissue damage in amebic
monocytes inhibit lymphocyte-dependent activation of intestinal infection of human intestine. Gastroenterology 1998; 115:1446–53.
epithelial cells by commensal bacteria. Microbiol Immunol 28 Zareie M, McKay DM, Kovarik GG, Perdue MH. Monocyte/
2002;46:195–205. macrophages evoke epithelial dysfunction: indirect role of tumor
12 Chantret I, Barbat A, Dussaulx E, Brattain MG, Zweibaum A. necrosis factor-alpha. Am J Physiol 1998;275:C932–9.
Epithelial polarity, villin expression, and enterocytic differentiation 29 Satake M, Watanabe H, Miyamoto Y, Shimizu M. Induction of
of cultured human colon carcinoma cells: a survey of twenty cell nitric oxide synthase and subsequent production of nitric oxide not
lines. Cancer Res 1988;48:1936–42. involved in interferon-gamma-induced hyperpermeability of
13 Hidalgo IJ, Raub TJ, Borchardt RT. Characterization of the human CACO-2 intestinal epithelial monolayers. Biosci Biotechnol Bio-
colon carcinoma cell line (CACO-2) as a model system for intestinal chem 2001;65:428–30.
epithelial permeability. Gastroenterology 1989;96:736–49. 30 Pahl HL. Activators and target genes of Rel/NF-kappaB transcrip-
14 Pinto M, Robine-Leon S, Appay M-D et al. Enterocyte-like differ- tion factors. Oncogene 1999;18:6853–66.
entiation and polarization of the human colon carcinoma cell line 31 Eckmann L, Jung HC, Schurer-Maly C, Panja A, Morzycka-
CACO-2 in culture. Biol Cell 1983;47:323–30. Wroblewska E, Kagnoff MF. Differential cytokine expression by
15 Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, human intestinal epithelial cell lines. regulated expression of inter-
Coffman RL. Two types of murine helper T cell clone. I. Definition leukin 8. Gastroenterology 1993;105:1689–97.
according to profiles of lymphokine activities and secreted proteins. J 32 Jung HC, Eckmann L, Yang SK et al. A distinct array of proin-
Immunol 1986;136:2348–57. flammatory cytokines is expressed in human colon epithelial cells in
16 Nathens AB, Rotstein OD, Dackiw AP, Marshall JC. Intestinal response to bacterial invasion. J Clin Invest 1995;95:55–65.
epithelial cells down-regulate macrophage tumor necrosis factor- 33 Hosokawa T, Kusugami K, Ina K et al. Interleukin-6 and soluble
alpha secretion: a mechanism for immune homeostasis in the gut- interleukin-6 receptor in the colonic mucosa of inflammatory bowel
associated lymphoid tissue. Surgery 1995;118:343–50. disease. J Gastroenterol Hepatol 1999;14:987–96.
17 Spottl T, Hausmann M, Kreutz M et al. Monocyte differentiation in 34 Noguchi M, Hiwatashi N, Liu Z, Toyota T. Secretion imbalance
intestine-like macrophage phenotype induced by epithelial cells. J between tumour necrosis factor and its inhibitor in inflammatory
Leukoc Biol 2001;70:241–51. bowel disease. Gut 1998;43:203–9.
18 Christ AD, Colgan SP, Balk SP, Blumberg RS. Human intestinal 35 Daig R, Rogler G, Aschenbrenner E et al. Human intestinal epithe-
epithelial cell lines produce factor(s) that inhibit CD3-mediated lial cells secrete interleukin-1 receptor antagonist and interleukin-8
T-lymphocyte proliferation. Immunol Lett 1997;58:159–65. but not interleukin-1 or interleukin-6. Gut 2000;46:350–8.
19 de Waal Malefyt R, Abrams J, Bennett B, Figdor CG, de Vries JE. 36 Haller D, Holt L, Parlesak A et al. Differential effect of immune cells
Interleukin 10 (IL-10) inhibits cytokine synthesis by human mono- on non-pathogenic Gram-negative bacteria-induced nuclear factor-
cytes: an autoregulatory role of IL-10 produced by monocytes. J Exp kappaB activation and pro-inflammatory gene expression in intest-
Med 1991;174:1209–20. inal epithelial cells. Immunology 2004;112:310–20.
20 Fiorentino DF, Zlotnik A, Mosmann TR, Howard M, O’Garra A. 37 Sadlack B, Merz H, Schorle H et al. Ulcerative colitis-like disease in
IL-10 inhibits cytokine production by activated macrophages. J mice with a disrupted interleukin-2 gene. Cell 1993;75:253–61.
Immunol 1991;147:3815–22. 38 Cario E, Becker A, Sturm A et al. Peripheral blood mononuclear
21 Chantry D, Turner M, Abney E, Feldmann M. Modulation of cells promote intestinal epithelial restitution in vitro through an
cytokine production by transforming growth factor-beta. J Immunol interleukin-2/interferon-gamma-dependent pathway. Scand J
1989;142:4295–300. Gastroenterol 1999;34:1132–8.

# 2004 Blackwell Publishing Ltd. Scandinavian Journal of Immunology 60, 477–485

You might also like