Sie sind auf Seite 1von 12

Veterinary Immunology and Immunopathology 111 (2006) 239250

www.elsevier.com/locate/vetimm

Effect of orally administered probiotic E. coli strain Nissle 1917


on intestinal mucosal immune cells of healthy young pigs
Swantje C. Duncker a,1, Axel Lorentz b, Bernd Schroeder a,
Gerhard Breves a, Stephan C. Bischoff b,*
a
Department of Physiology, School of Veterinary Medicine, Bischofsholer Damm 15, 30173 Hannover, Germany
b
Department of Nutritional Medicine and Prevention, University of Hohenheim, Fruwirthstr. 12, 70593 Stuttgart, Germany
Received 8 August 2005; received in revised form 8 December 2005; accepted 16 January 2006

Abstract

Several beneficial effects of probiotics have been described in studies using rodent disease models and in human patients;
however, the underlying mechanisms remained mostly unclear. Only a few studies focused on the effects of probiotics on the
intestinal mucosal immune system. Here, we studied the effect of the probiotic strain E. coli Nissle 1917 (EcN) administered
orally to young pigs at two concentrations (109 and 1011 CFU/d for 21 days) on the gut-associated lymphatic tissue. This
probiotic strain was shown recently to reduce recurrence of inflammation in ulcerative colitis patients. We quantified the number
and distribution of intestinal immune cells (granulocytes, mast cells, CD4+, CD8+, CD25+, IgA+ lymphocytes) and the mucosal
mRNA expression of cytokines (IFN-gamma, TNF-alpha, TGF-beta, IL-10) and antimicrobial peptides (PR-39, NK-lysin,
prepro-defensin-beta 1, protegrins). The number and distribution of cells were highly different between small intestinal and
colon segments in all groups, but were not influenced by EcN, except high dose EcN fed pigs (1011 CFU/d) showing an increase
in mucosal CD8+ cells in the ascending colon. The mRNA analysis revealed no changes associated with EcN feeding. In
conclusion, according to our analyses EcN has only minor effects on the distribution of mucosal immune cells in the gut of
healthy individuals. The well-established preventive effects of EcN might therefore be relate to other mechanisms than simple
modulation of immune cell distribution.
# 2006 Elsevier B.V. All rights reserved.

Keywords: Probiotics; Intestine; Porcine; GALT; Lymphocytes; E. coli Nissle 1917; CD8

Abbreviations: AMP, anti-microbial peptide; CD, cluster of differentiation; EcN, Escherichia coli Nissle 1917; GALT, gut associated
lymphoid tissue; IEL, intraepithelial lymphocytes; LP, lamina propria; UC, ulcerative colitis
* Corresponding author. Tel.: +49 711 459 4101; fax: +49 711 459 4343.
E-mail addresses: duncker@mcmaster.ca (S.C. Duncker), bischoff.stephan@uni-hohenheim.de (S.C. Bischoff).
1
Present address: Brain-Body Institute (BBI), 50 Charlton Ave. E, Hamilton, Ont., Canada L8N 4A6. Tel.: +1 905 522 1155x2277/5284.

0165-2427/$ see front matter # 2006 Elsevier B.V. All rights reserved.
doi:10.1016/j.vetimm.2006.01.017
240 S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250

1. Introduction ulcerative colitis (UC) in humans (Kruis et al., 1997,


2004; Rembacken et al., 1999). The mechanisms
Particular probiotic strains have been successfully underlying the beneficial effects of EcN are unclear
used for prophylaxis of intestinal infection in live- as yet, immunomodulating effects have been
stock animals (Vanbelle et al., 1990; Alexopoulos discussed (Hockertz, 1997; Schultz et al., 2004;
et al., 2004). In humans, they are known to prevent Sturm et al., 2005). Here, we aimed to further
atopic dermatitis (Lodinova-Zadnikova et al., 2003; elucidate the mechanisms of action in an animal
Kalliomaki et al., 2003), gastrointestinal infectious system thought to have a rather similar intestinal
and antibiotic associated diarrhoea (McFarland et al., anatomy and structure of the mucosal immune
1995; DSouza et al., 2002), as well as inflammatory system compared to man. This seems to be of
bowel disease such as ulcerative colitis (Kruis et al., relevance, since beneficial clinical effects of EcN
1997, 2004; Rembacken et al., 1999). Furthermore, were only shown in humans and calves (von Buenau
some probiotics such as Lactococcus lactis have been et al., 2005) so far. Therefore, we chose pigs for our
used for delivery of cytokines towards the intestinal ex vivo studies and examined the effects of EcN on
mucosa (Steidler et al., 2003). Even though selective immune cell distribution and cytokine expression as
probiotics such as Lactobacillus GG (Kalliomaki well as AMP (antimicrobial peptides) expression in
et al., 2001, 2003), Saccharomyces boulardii (McFar- the intestinal mucosa.
land et al., 1995) and E. coli Nissle 1917 (Kruis et al.,
1997, 2004; Rembacken et al., 1999) have been
proven to be clinically effective, the modes of action 2. Material and methods
by which they achieve their beneficial effects
remained unclear. In vitro experiments suggested 2.1. Animals
that both modulation of the gut flora and of the
intestinal cytokine production subsequently leading Fifteen young adult pigs with a mean weight of
to modulation of the adaptive immune response might 16  2.7 kg were used for experiments. The pigs
be of relevance (Christensen et al., 2002; Vaarala, originated from different litters of a conventional
2003). So far, only a few studies have investigated the crossbred program of German Landrace and
influence of probiotics on the number and distribution Pietrain carried out by the university research
of intestinal immune cells in vivo (Perdigon et al., farm. All animals were on a regular deworming
1995; Pestka et al., 2001). schedule with 0.3 mg/kg ivermectin two times a year.
The probiotic E. coli strain Nissle 1917 (EcN) Upon their arrival the pigs were housed on straw.
used in this study is of the serotype O6:K5:H1 and They were maintained on a regular mixed diet for
was isolated in 1916 by the German physician Alfred growing pigs. Following a 2 weeks adaptation period
Nissle (Loew, 2000). Since then this bacterial strain the animals were divided into three groups of five pigs
has been used as a probiotic drug and is considered each. The first group served as control. The second
to be safe (Blum et al., 1995; Grozdanov et al., 2002, group received 109 CFU EcN/d and the third group
2004; Westendorf et al., 2005). Oral administration 1011 CFU EcN/d. EcN was given as a suspension in
of up to 7.2  109 CFU of EcN per animal did not dilution medium (2.5 g NaCl, 2.5 g KCl, 0.2 g
cause colitis or urogenital infection in neonatal MgSO4, 0.2 g CaCl2, 0.2 g MgCl26H2O, 20 ml
gnotobiotic piglets (Gunzer et al., 2002). EcN NaOH (32%) add distilled H2O to a total of
contains different so-called fitness factors such 1000 ml) orally for 21 days. The control group
as siderophores and microcins (Patzer et al., 2003; received the dilution medium without EcN. The
Grozdanov et al., 2004). Moreover, the bacterium animals were killed by exsanguination following
shows a distinct LPS-structure supposed to exert mechanical stunning. The abdominal contents were
particular immune modulating properties (Grozda- removed immediately after death. The protocol of
nov et al., 2002). EcN is the active component of animal treatment was approved and the procedure
Mutaflor1, a drug showing equivalent effectiveness was supervised by an animal protection officer of the
as 50 -amino-salicylates in maintaining remission in University.
S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250 241

2.2. Fecal samples 2.4. Histology

To ensure proper administration of EcN, fecal Paraformaldehyde-fixed paraffin slides were


samples were taken from every animal at days 7 deparaffinized, hydrated and stained using Meyers
and 14 of the experiment and after slaughter. They hemalum solution (Merck, Darmstadt, Germany) and
were analyzed for EcN content by multiplex PCR aqueous eosin Y Solution (SigmaAldrich, Stein-
using strain-specific DNA primer sequences as heim, Germany) according to standard protocols
described elsewhere (Blum-Oehler et al., 2003). (Romeis, 1989). Carnoys solution fixed paraffin
All EcN-treated animals were tested positive for sections were stained with toluidine blue O (Sigma
EcN at days 7 and 14 whereas control animals Aldrich) for an hour and counterstained with Meyers
always yielded negative EcN RT-PCR results (data haemalum. Following dehydration with increasing
not shown). concentrations of alcohol and finally xylene, the
slides were mounted in Entellan1 (Merck).
2.3. Tissue samples
2.5. Monoclonal antibodies
Tissue samples were taken immediately after
slaughter from the duodenum (10 cm aboral from The antibodies used in this study are listed in
the pylorus), jejunum (4.50 m aboral from the Table 1. Biotinylated secondary Ab (sAb) were
pylorus), ileum (5 cm oral from the ileocolic obtained from Southern Biotechnology, Birmingham,
orifice), ascending (10 cm aboral from the caeco USA.
colic junction) and descending (60 cm aboral from
the end of the ascending colon) colon. Mucosa 2.6. Immunohistochemistry
preparations and small tissue specimen (0.5 cm)
were immediately snap frozen in liquid nitrogen for Different immune cell populations were identified
RNA isolation and cryostat sectioning, respectively. by immunohistochemistry using the enzyme-linked
An additional tissue specimen was collected at each immunoperoxidase technique. Incubation steps were
location (0.5 cm) and fixed either in 4% parafor- carried out in a humid chamber at room temperature
maldehyde at pH 7.2 for 1824 h or in Carnoys if not indicated otherwise. After each step slides were
solution (60% ethanol absolute, 30% chloroform, washed for 5 min in TBS (pH 7.4) supplemented with
10% glacial acetic acid) for 2 h. The samples were 0.035% Tween120 (SigmaAldrich). CD4+ and
embedded in paraffin wax and sectioned at 2 mm, CD8+ subpopulation analysis was performed using
placed on SuperfrostTM plus slides (Menzel, cryostat sections of 5 mm thickness placed on
Braunschweig, Germany) and dried overnight at Superfrost1 plus glass slides (Menzel-Glaeser,
37 8C. Braunschweig, Germany). Sections were air-dried

Table 1
Antibody dilutions
Primary Ab Clone Company Dilution Secondary Ab
a
CD4 74-12-4 VMRD, USA 1/50 Goatanti-mouse IgG2b
CD8 MIL-12 Serotec, UK 1/100 b Goatanti-mouse IgG2a
CD25 231.3B2 Serotec, UK 1/20a Goatanti-mouse IgG1
IgA K61 1B4 Serotec, UK 1/10a Goatanti-mouse IgG1
Mouse IgG1 Dianova, Germany 1/20a Goatanti- mouse IgG1
Mouse IgG2a Dianova, Germany 1/100 c Goatanti-mouse IgG2a
Mouse IgG2b Dianova, Germany 1/50b Goatanti-mouse IgG2b
a
Dilutions were prepared with Monet Blue (Biocare, Germany).
b
Dilutions were prepared with VanGogh Yellow (Biocare, Germany).
242 S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250

for 30 min and fixed in a mix of equal amounts of compartment of the mucosa was considered to be the
acetone and methanol at 4 8C. Tissues were blocked villi in the small intestine and the most luminal
with 5% normal swine serum diluted in Tris-buffered microscopic field of view (1000 times magnification)
saline (TBS) for 30 min to reduce unspecific Ab- in the large intestine. The basal compartment of the
binding, and then incubated overnight with the mucosa contained the crypts in the small intestine and
primary Ab at 4 8C. The following day slides were the most basal microscopic field of view in the large
blocked again with 5% normal swine serum diluted in intestine. The assignment made in the large intestine
TBS, and then incubated with a biotinylated ensured that cells were not accidentally counted for
secondary Ab (1/50 dilution) for 30 min. Streptavi- both compartments. CD4+ cells could be evaluated on
dinhorse radish peroxidase (Biocare, Hamburg, a semi-quantitative basis only by assignment to four
Germany) and 3-amino-9-ethyl carbazol (AEC) were groups defined as follows: (1) <25% of LP cells
used to visualize specific binding of the Ab (Zymed, CD4+; (2) 2549% of LP cells CD4+; (3) 5075% of
San Francisco, CA). Slides were counterstained with LP cells CD4+; (4) >75% of LP cells CD4+.
Meyers hemalum solution for improved cell differ-
entiation and finally mounted in Faramount1 Aqu- 2.8. RNA isolation and RT-PCR
eous Mounting Medium (Dako, Hamburg, Germany).
Each staining series was accompanied by one slide Total RNA was prepared from washed and stripped
treated with an isotype control replacing the primary mucosa samples (0.90.8 g) using the RNeasy Mini
Ab. Because of the lack of endogenous peroxidase Kit (Qiagen, Hilden, Germany) following the manu-
activity in the cryostat sections, blocking with facturers manual. The amount of total RNA was
hydrogen peroxide was not necessary using this quantified by spectrometry. For RT-PCR 200 ng of
staining protocol. total RNA was used and processed as described
For the detection of CD25 and IgA paraffin previously (Gebhardt et al., 2002). Briefly, RNA was
embedded specimen were used. Slides were depar- treated with 10 U RNase-free DNAse (Promega,
affinized and hydrated. To ensure antigen de-masking Madison, WI) to remove genomic DNA. After
the samples were treated with Dako1 Target Retrival denaturation at 70 8C cDNA was synthesized at
Solution (pH 6) (Dako) in a water bath at 95 8C. The 49 8C adding SuperscriptTM plus reverse transcriptase
subsequent immunohistochemistry was performed as (Invitrogen, Karlsruhe, Germany) and oligo-dT
described above; however, an additional incubation in primers (Pharmacia, Uppsala, Sweden). 1/10 of the
methanol containing 1.2% hydrogen peroxide was total cDNA was used for one PCR reaction. PCR was
added following the incubation with the sAb to ensure performed with 30 s at 94 8C, 45 s at 5558 8C, 35 s at
quenching of endogenous peroxidase. 72 8C. Numbers of cycles and annealing temperatures
differed for each pair of primers: IFN-gamma (34
2.7. Cell counts cycles, annealing 58 8C), TNF-alpha (35 cycles,
annealing 58 8C), TGF-beta (32 cycles, annealing
Cells counts within the intestinal mucosa were 58 8C), IL-10 (40 cycles, annealing 58 8C) prepro-
performed at 1000-fold magnification using a light defensin-beta 1 (39 cycles, annealing 58 8C), prote-
microscope Labophot-2 (Nikon, Tokyo, Japan). Cells grins and PR-39 (42 cycles, annealing 55 8C), NK-
were counted separately in the epithelium and the lysin (36 cycles, annealing 55 8C) and GAPDH (28
lamina propria (LP). Data were expressed in percent of cycles, annealing 56 8C). To ensure that a saturation of
positive cells per total number of epithelial cells or primers were not misinterpreted as an increase of
total number of cells in the LP, respectively. Cells were mRNA, as many cycles of PCR were used as were
assigned to the intraepithelial compartment only if necessary to ensure the amplification reaction was still
they were located with the epithelial cell band or at in its exponential phase. The number of cycles for
least half of their plasma membrane was in contact every primer pair was determined by preliminary
with the basement membrane of the epithelium. For experiments with samples from at least three different
CD8+ cells, we differentiated between luminal and untreated pigs and for every intestinal location tested.
basal section of the mucosa. Therefore, the luminal The cycle number was than used in treated and
S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250 243

Table 2
PCR-primer
Sense-primer Antisense-primer
IFN-gamma 50 -GCAAGTACCTCAGATGTACC-30 50 -TGGCCTTGGAACATAGTCTG-30
TNF-alpha 50 -ACTGCACTTCGAGGTTATCG-30 50 -AGAGGACCTGGGAGTAGATG-30
TGF-beta 50 -CAGAGAGGCTATAGAGGGTT-30 50 -TGTCTAGGCTCCAGATGTAG-30
IL-10 50 -TTGCCAAGCCTTGTCAGAGA-30 50 -TCACCCATGGCTTTGTAGAC-30
PR-39 50 -ACCCATCCATTCACTCAC-30 50 -AGCCACAACAATAAGATCC-30
NK-lysin 50 -GAGCAGTTCTGCCAGAACCT-30 50 -GCAGGAGTTAGGTGAGAGAA-30
Protegrines 50 -TGGATCAGATCAAGGACC-30 50 -ACACAGACGCAGAACCTAC-30
Prepro-defensine-beta 50 -CCTCCTTGTATTCCTCCTCA-30 50 -TTGCAGCATTTGACTTGGGG-30
GAPDH 50 -ACCACAGTCCATGCCATCAC-30 50 -TCCACCACCCTGTTGCTGTA-30

untreated animals to allow comparative semi-quanti- significantly different when comparing small and
tative detection of mRNA expression. PCR was large intestinal sites, their density within the LP
performed with 2.5 U Taq DNA polymerase (Invitro- ranging from 0.4% in the jejunum to 3.7% in the
gen) and 20 pmol of specific sense and antisense duodenum and colon (Fig. 1B). Mast cells were
primers shown in Table 2. The PCR products equally distributed throughout the intestinal LP,
contained cDNA fragments of 359 bp (IFN-gamma), and accounted for about 3% of LP cells at all
266 bp (TNF-alpha), 371 bp (TGF-beta), 243 bp
(IL-10), 168 bp (prepro-defensin-beta 1), 262 bp
(PR-39), 100 bp (protegrins), 479 bp (NK-lysin)
and 452 bp (GAPDH), respectively. Eleven micro-
liters of the PCR products were visualized on 1%
agarose gel containing ethidium bromide (500 ng/ml)
and photographed.

2.9. Statistics

Data were expressed as mean (S.D.) if not


indicated otherwise. Differences in cell counts were
accessed using the Students t-test and differences
between experimental groups by one-way ANOVA.

3. Results

3.1. Number and distribution of granulocytes and


mast cells

In untreated animals, granulocytes and mast cells


were detected in the lamina propria of the mucosa, but
not in the epithelium. Eosinophils were concentrated
within the basal part of the LP. They were much more
frequent in the small compared to the large intestine, Fig. 1. Eosinophil (A), neutrophil (B) and mast cell (C) density in
with highest numbers in the mucosa of the ileum the intestinal lamina propria mucosae (LP) shown as per cent of all
lamina propria cells. Animals were either untreated (white bars), or
(11  2.2% of total LP cells). In the colon almost no treated with 109 (hatched bars) or 1011 CFU/d (black bars) of E. coli
eosinophils were visible (0.6  0.3% of total LP Nissle 1917 (EcN). Bars show the mean of five pigs per group with
cells) (Fig. 1A). Neutrophil numbers were not error bars indicating the standard deviation.
244 S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250

investigated sites of the gut (Fig. 1C). No basophils the more caudal sites of the GI tract (jejunum, ileum,
could be detected in the intestinal mucosa of the pigs. colon), where only 3% of LP cells were IgA plasma
The numbers of the three cell types examined here cells. The density of IgA plasma cells was not
did not change following treatment of the animals changed by treatment of the animals with EcN
with either low or high dose of EcN, as shown in (Fig. 2A).
Fig. 1. Cells expressing the CD4 co-receptor were
frequently found in the LP but were absent in the
3.2. Number and distribution of T lymphocyte epithelium. Within the LP 5075% of cells were CD4
subsets and plasma cells positive. Therefore, we only performed a semi-
quantitative analysis. CD4+ cells were evenly
IgA producing plasma cells, CD4+ cells, CD25+ distributed throughout cross-sections of the mucosa
cells and CD8+ cells were analyzed by immunohis- in the ileum and colon; however, the mucosa of the
tochemistry. IgA plasma cells were predominantly duodenum and jejunum contained higher numbers of
found in the LP of the crypts. They were virtually CD4+ cells in the LP of the villi compared to the
absent in the epithelium and in submucosal layers. crypts (Fig. 2B). CD25+ cells were localized in small
Number of IgA plasma cells were highest in the amounts (2.43.7% of total LP cells) in the intestinal
duodenum with a density of 21.1% (3.1% of LP mucosa. They were evenly distributed in the LP
cells). Plasma cell counts were significantly lower at (Fig. 2C) and only sporadically detected in the
epithelium. Again, neither CD4+ cell density nor
CD25+ cell density changed following EcN treatment
(Fig. 2B and C).
CD8+ cells were the only ones found to be
consistently present within the epithelium and the LP
of the intestinal mucosa. The density of CD8+ cell
was higher in the luminal compared to the basal

Fig. 3. Density of CD8+ cells in the luminal compartment of the


mucosa (A) and the basal compartment of the mucosa (B), shown in
percent of all epithelial cells, LP cells, respectively. Animals were
Fig. 2. Density of IgA+ plasma cell (A), CD4+ cells (B) and CD25+ either untreated (white bars), or treated with 109 (hatched bars) or
cells (C) in the intestinal lamina propria mucosae (LP) shown as per 1011 CFU/d (black bars) of E. coli Nissle 1917 (EcN). First bar of
cent of all lamina propria cells. Animals were either untreated (white every colour pattern indicating cells in the epithelium and the second
bars), or treated with 109 (hatched bars) or 1011 CFU/d (black bars) bar cells in the LP. Bars show the mean of five pigs per group with
of E. coli Nissle 1917 (EcN). Bars show the mean of five pigs per error bars indicating the standard deviation. *p < 0.05 compared to
group with error bars indicating the standard deviation. same intestinal segment in the control group.
S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250 245

compartment of the mucosa. Highest counts were (1011 CFU/d) showed significantly higher CD8+
found in the luminal epithelium of the duodenum. cells counts in the luminal LP of the Ileum, in the
Along the intestinal mucosa the CD8+ cell counts epithelium and the lamina propria of the ascending
in the LP were reverse to those in the epithelium colon compared to placebo-fed pigs (Figs. 3 and 4).
of the same compartment. For example in the In low dose fed animals CD8+ cells where increased
luminal compartment of the mucosa the epithelium in the basal epithelium of the jejunum and in the
contained much higher numbers of CD8+ cells than luminal LP of the ascending colon (Fig. 3). No
the corresponding LP (Fig. 3A). In the basal significant changes in CD8+ cell counts were obse-
compartment of the mucosa the opposite was true rved in the mucosa of other intestinal segments on
(Fig. 3B). Animals receiving high amounts of EcN EcN treatment.

Fig. 4. Pictures showing cells bearing the CD8 antigen (red) in the mucosa of the ascending colon. The animal treated with 1011 CFU/d (B) of
EcN shows an increase in the number of CD8 positive cells compared to the untreated animal (A). Slides were stained by immunohistochemistry
using the MIL-12 Ab (Serotec, UK) or a mouse IgG2a (Dianova, Germany) isotype as negative control (C). Pictures from one representative
animal per group are shown. Metering bar = 25 mm.

Fig. 5. mRNA expression of cytokines (IFN-gamma, TNF-alpha, TGF-beta, IL-10) and antimicrobial peptides (prepro-defensin-beta 1, NK-
lysin) in the ileum and the ascending colon compared to housekeeping gene (GAPDH). Pictures show two representative animals of the control
group and the group treated with 1011 CFU/d of E. coli Nissle 1917 (EcN). Numbers indicate the PCR cycles accomplished.
246 S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250

3.3. mRNA expression of different cytokines system is considered to be much closer to the human
and AMP one than the rodent immune system (Miller and
Ullrey, 1987; Boes and Helwigh, 2000). Studies have
Total RNA from intestinal mucosa of duodenum, been performed in pigs on the distribution of
ileum and ascending colon in control animals and particular immune cells in the blood and in different
animals fed 1011 CFU EcN/d was investigated. tissues (Bianchi et al., 1992; Yang and Parkhouse,
mRNA expression of different cytokines and anti- 1996; Boeker et al., 1999; Saalmuller et al., 2002;
microbial peptides was evaluated semi-quantitatively Sinkora et al., 2005) including studies on T cells in the
by comparison to GAPDH as housekeeping gene to intestine (Vega-Lopez et al., 1993; Pabst and
check for possible changes in immune cell sub- Rothkotter, 1999). Nevertheless, it is surprising to
populations. mRNA expression of IFN-gamma, TNF- note that no published data are available on the
alpha, TGF-beta and IL-10 as well as the mRNA distribution of cells of the innate immune system
expression of the AMP: prepro-defensine-beta 1 and within the intestine of the pig. Therefore, we were
NK-lysin was detected in the duodenum (data not interested not only in studying the influence of EcN on
shown), ileum and ascending colon (Fig. 5). In all intestinal immune cells, but also in quantifying the
samples PR-39 was only partly seen in the intestinal number and distribution of granulocytes, mast cells
mucosa and mRNA for protegrins was absent in all and lymphocyte subsets in the gut mucosa.
animals and intestinal segments (data not shown). Eosinophils were mainly found in the LP of the
Transmural duodenal tissue samples of 3-week-old crypts in the small intestine and the basal part of the
piglets were included as positive controls. Neither in crypts in the large intestine, as described previously
the investigated cytokines nor in the AMP changes (Bianchi et al., 1992). Eosinophils could be found at
were detected after feeding high amounts of EcN all sites of the small and large intestine, the highest
(1011 CFU/d). numbers being seen in the ileum. Interestingly, we
found for the first time that the density of eosinophils
in the colon LP was lower than 1% and thus much
4. Discussion lower than that described in humans (Bischoff et al.,
1996). Because none of the animals showed signs of
During the last decades pigs health became a huge parasite infection and all were dewormed on a routine
economic factor in livestock farming because of schedule, an increase of eosinophils in the small
intensive animal husbandry and increasing produc- intestine due to parasitosis can be largely ruled out.
tion rates. Simultaneously, the opportunities of There is evidence from the literature that gastro-
prophylactic treatment against the major porcine intestinal eosinophils not only act as inflammatory
pathogens were limited due to governmental restric- effector cells of allergy and parasitic infection, but are
tion (prohibition of antibiotics in animal feed, etc.). also capable of functioning as immunomodulating
Therefore, alternatives are needed. Probiotics showed cells (Kato et al., 1998). Kato et al. showed a
the potential to fulfil such requirements, even though spontaneous minimal degranulation of intestinal
the mechanisms of their beneficial effects are mostly eosinophils in the healthy gut that does not occur in
unknown. This study was performed to investigate the other normal tissues. These data suggest that
effects of EcN, a probiotic E coli strain successfully eosinophils may also act as immune-modulating cells
used in human and veterinary medicine, on the in the small intestine of pigs.
intestinal immune system of healthy young pigs. The Compared to eosinophils the amount of neutro-
aim was to further understand the modes of action of phils in the intestinal mucosa of healthy pigs was
this probiotic strain, which obviously has beneficial much lower. Neutrophil counts were highest in the
properties both in man (Kruis et al., 2004) and in duodenum and descending colon. The amount of
calves (von Buenau et al., 2005). We selected the pig neutrophils was higher than the numbers regarded
as a model not only because this species might as well to be normal in humans (0.4%) (Ogra et al., 1999). In
benefit of probiotic treatment (Alexopoulos et al., the duodenum the reason might be that pigs are
2004), but also because the pig intestinal immune more prone to contact with microorganisms because
S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250 247

of their usual environment. Therefore, more neutro- other probiotics like Saccharomyces boulardii (Buts
phils may ensure a fast reaction to potential et al., 1990; Rodrigues et al., 2000).
pathogens. The relatively high amount of neutrophils Heterogeneous results were obtained with regard to
in the colon, compared to humans, is likely to be a lymphocyte subsets. We found no changes in CD4+
result of higher physiologic amounts of bacteria in and CD25+ cells, but an increase of CD8+ cells in the
this part of the pig intestine compared to the human ascending colon induced by EcN. In contrast to the
colon. In contrast to neutrophils, the counts of mast results of others (Vega-Lopez et al., 1993), we found in
cells were in the expected ranges, since our data were our experiments polarization of mucosal CD4+ cells
similar to those reported by Xu et al. (1993) who did towards the LP of the villi only in the duodenum and
similar studies in pigs. Mast cells, unlike granulo- jejunum. In the ileum and colon, the CD4+
cytes, were evenly distributed throughout the intest- lymphocytes were equally distributed throughout
inal segments. the mucosa. An explanation of this difference might
Treatment with EcN had no effect on the number be the lower age of the animals used in the study by
and distribution of granulocytes and mast cells in Vega-Lopez et al. Even though no alterations in the
healthy pigs. Moreover, the absence of major changes CD4+ cell counts were evident between treatment and
in tested cytokine and AMP mRNA expression makes control groups, an influence EcN on the balance of
an influence of EcN on the cellular metabolism CD4+ subpopulations cannot be ruled out. Different
unlikely too. Even though, a definite conclusion on from CD4+ cells significantly more CD8+ cells were
cytokine-mRNA concentration would require a quan- counted in the luminal epithelium than in the lamina
titative method for mRNA detection like real time propria of the intestine, as reported earlier by other
PCR, because the semi-quantitative method used can groups (Vega-Lopez et al., 1993, 2001; Pabst and
only detect major changes. However, the unaltered Rothkotter, 1999). This holds true for all intestinal
number of cells in healthy EcN-treated animals does segments investigated. We extended these findings by
not rule out a positive effect on disease-induced cell distinguishing between the epithelium and the LP in
recruitment (Sturm et al., 2005; Kamada et al., 2005). the luminal and the basal part of the mucosa,
From experiments with other probiotics like lactoba- respectively. The highest number of CD8+ cells
cilli it is known, that they are able to decrease (one-third of total cell count) was found in the luminal
infection-induced, transmucosal migration of neutro- epithelium of the duodenum. Different from the
phils (Michail and Abernathy, 2002). Keeping in mind findings of Vega-Lopez et al. we could not confirm that
EcN being a probiotic, the fact that it does not recruit the CD8+ cells of the LP were localized near the
neutrophils in healthy animals rather underlines its basement membrane of the epithelium, possibly
apathogenicity than its ineffectiveness. Even though in because of different methodological approaches. We
the present study no changes were detected with report here for the first time that EcN treatment
respect to the number and distribution of intestinal increased the amount of CD8+ cells in the ascending
eosinophils, it still needs to be clarified if the colon in healthy young pigs. Feeding 1011 CFU EcN/d
administration of EcN leads to alterations in mediator for 21 days resulted in an increase of CD8+ cells in the
composition and release in this cells. ascending colon, both in the luminal as well as in the
In accordance with findings of other authors basal compartment of the mucosa. In the absence of
(Rothkotter et al., 1991; Bianchi et al., 1992) IgA- mucosal inflammation (no increase in neutrophils, no
producing plasma cells were mainly found in the basal differences of proinflammatory cytokine-mRNA
part of the mucosa with highest amounts in the crypts expression in comparison between treated and
of the duodenum. No changes were detected following untreated groups, no activation of lymphocytes) the
EcN treatment. An increase in the number of intestinal high numbers of CD8+ cells cannot be interpreted as
IgA producing plasma cells, does not seem to be a an inflammatory response. From our experiments it
mechanism of action of EcN in conventionally reared, cannot be concluded whether the increased number of
healthy pigs. Nevertheless, unchanged IgA plasma CD8+ cells detected in the epithelium of the ascending
cell counts do not rule out an influence of EcN on the colon is due to proliferation of CD8+ cells or to
luminal secretory IgA concentration as reported for recruitment of cells from other sites like the lamina
248 S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250

propria or the mesenteric lymph nodes. However, it directly fluorescent-labeled antibodies to perform
has been shown in mice that intestinal IEL are able to fluorescent immunohistochemistry and an improve-
proliferate in the epithelium (Lin et al., 1994). The ment in isolation techniques for intestinal immune
accumulation of lymphocytes originating from the cells will be important to defining specialized subsets
peripheral blood as an explanation of the increase of of lymphocytes and innate immune cells in situ. This
CD8+ cells due to EcN treatment is unlikely, since will help to further investigate an influence of EcN on
Sturm et al. (2005) showed a down-regulation of intestinal immune cells in the pig. Furthermore,
proliferation in peripheral blood T cells by EcN in using EcN in an infection model might be useful to
vitro, whereas the proliferation of LP T cells was reveal changes in the reaction and migration of
unchanged. The cell population expressing CD8 co- intestinal immune cell population that cannot be
receptor in the intestine consists of different cell types detected in healthy animals.
including cytotoxic T cells, gamma/delta T cells and
NK cells. In this study we did not differentiate
between subtypes of CD8+ cells as CD8high and 5. Conclusions
CD8low. Therefore, no conclusion can be drawn on
changes within a special subtype of CD8+ cells; We showed in this study as described earlier in
however, the beneficial effects of probiotics such as humans (Kruis et al., 2004) and mice (Waidmann
EcN could be related to known effects of CD8+ et al., 2003; Schultz et al., 2004), that EcN did not
subpopulations, such as defence against pathogens by induce any signs of inflammation and did not exhibit
cytotoxic CD8+ cells, or maintenance and enhance- any signs of pathogenicity, even when fed to young
ment of the epithelial integrity by gamma/delta CD8+ pigs at high doses. The EcN-induced enhancement of
cells (Chen et al., 2002). Other than in humans and the number of CD8+ cells in the ascending colon
mice intraepithelial CD8+ gamma/delta T cells have might be linked to the beneficial role of maintaining
not yet been identified in the intestine of pigs, but that porcine intestinal health and preventing disease.
does not rule out the possibility that equivalent Intestinal epithelial cells have been shown in vitro to
subtypes of CD8+ cells exist also in pigs within the produce keratinocyte growth factor (KGF) (Chen
epithelium. Especially, because there is increasing et al., 2002) and might thereby increase the epithelial
evidence that pigs have a distinct repertoire of integrity. Mice lacking gamma/delta TCR IEL have
intestinal gamma/delta TCR (Holtmeier et al., 2002) impaired epithelial development in the intestine
and T cells expressing the delta chain of the TCR have (Boismenu and Havran, 1994). It has also been
been shown in the colonic epithelium (Hontecillas shown recently that IEL express junctional molecules
et al., 2005). Future experiments are needed to further like epithelial cells, whereas lymphocytes from other
characterize the phenotype of CD8+ cells in the pig, body sites (spleen, mesenteric lymph nodes, thymus,
and to establish their biological relevance in disease Payers patches) do not (Inagaki-Ohara et al., 2005).
and disease prevention. This opens up new possibilities for the involvement of
Even though the porcine intestinal immune IEL in the epithelial barrier and for the communica-
system might be an excellent model in terms of tion with epithelial cells. To further evaluate the
transfer of results to the situation in humans, there are beneficial effects of EcN in pigs and the underlying
of course some limitations. The lack of EcN effects mechanisms, experiments with pig infection models
on the density of innate immune cells such as are of special interest.
granulocytes or mast cells shown in this study, as
well as on the density of CD4+ cells (mostly T helper
lymphocytes), CD25+ (activated T cells) and IgA Acknowledgements
cells might be the result of the limited number of
animals examined here. Unfortunately, the repertoire We thank Gisela Weier and Gerhild Becker for
of antibodies directed against porcine cell surface excellent technical assistance and Yvonne Armbrecht
antigens is limited compared to that available for and Michael Rohde for help with the animals as well
rodents. A future improvement in availability of as Anne Luschert for advice in immunohistochem-
S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250 249

istry. This work was supported by Ardeypharm cing growth and IgE receptor-dependent mediator release. Eur. J.
Immunol. 32, 23082316.
GmbH, Herdecke, Germany and the Deutsche For-
Grozdanov, L., Raasch, C., Schulze, J., Sonnenborn, U., Gottschalk,
schungsgemeinschaft (SFB 280). G., Hacker, J., Dobrindt, U., 2004. Analysis of the genome
structure of the nonpathogenic probiotic Escherichia coli strain
Nissle 1917. J. Bacteriol. 186, 54325441.
References Grozdanov, L., Zahringer, U., Blum-Oehler, G., Brade, L., Henne,
A., Knirel, Y.A., Schombel, U., Schulze, J., Sonnenborn, U.,
Alexopoulos, C., Georgoulakis, I.E., Tzivara, A., Kyriakis, C.S., Gottschalk, G., Hacker, J., Rietschel, E.T., Dobrindt, U., 2002. A
Govaris, A., Kyriakis, S.C., 2004. Field evaluation of the effect single nucleotide exchange in the wzy gene is responsible for the
of a probiotic-containing Bacillus licheniformis and Bacillus semirough O6 lipopolysaccharide phenotype and serum sensi-
subtilis spores on the health status, performance, and carcass tivity of Escherichia coli strain Nissle 1917. J. Bacteriol. 184,
quality of grower and finisher pigs. J. Vet. Med. A Physiol. 59125925.
Pathol. Clin. Med. 51, 306312. Gunzer, F., Hennig-Pauka, I., Waldmann, K.H., Sandhoff, R., Grone,
Bianchi, A.T., Zwart, R.J., Jeurissen, S.H., Moonen-Leusen, H.W., H.J., Kreipe, H.H., Matussek, A., Mengel, M., 2002. Gnotobiotic
1992. Development of the B- and T-cell compartments in piglets develop thrombotic microangiopathy after oral infection
porcine lymphoid organs from birth to adult life: an immuno- with enterohemorrhagic Escherichia coli. Am. J. Clin. Pathol.
histological approach. Vet. Immunol. Immunopathol. 33, 201 118, 364375.
221. Hockertz, S., 1997. Augmentation of host defence against bacterial
Bischoff, S.C., Wedemeyer, J., Herrmann, A., Meier, P.N., Traut- and fungal infections of mice pretreated with the non-pathogenic
wein, C., Cetin, Y., Maschek, H., Stolte, M., Gebel, M., Manns, Escherichia coli strain Nissle 1917. Arzneimittelforschung 47,
M.P., 1996. Quantitative assessment of intestinal eosinophils and 793796.
mast cells in inflammatory bowel disease. Histopathology 28, 1 Holtmeier, W., Kaller, J., Geisel, W., Pabst, R., Caspary, W.F.,
13. Rothkotter, H.J., 2002. Development and compartmentalization
Blum, G., Marre, R., Hacker, J., 1995. Properties of Escherichia coli of the porcine TCR delta repertoire at mucosal and extraintest-
strains of serotype O6. Infection 23, 234236. inal sites: the pig as a model for analyzing the effects of age and
Blum-Oehler, G., Oswald, S., Eiteljorge, K., Sonnenborn, U., microbial factors. J. Immunol. 169, 19932002.
Schulze, J., Kruis, W., Hacker, J., 2003. Development of Hontecillas, R., Bassaganya-Riera, J., Wilson, J., Hutto, D.L.,
strain-specific PCR reactions for the detection of the probiotic Wannemuehler, M.J., 2005. CD4+ T-cell responses and distribu-
Escherichia coli strain Nissle 1917 in fecal samples. Res. tion at the colonic mucosa during Brachyspira hyodysenteriae-
Microbiol. 154, 5966. induced colitis in pigs. Immunology 115, 127135.
Boeker, M., Pabst, R., Rothkotter, H.J., 1999. Quantification of B, T Inagaki-Ohara, K., Sawaguchi, A., Suganuma, T., Matsuzaki, G.,
and null lymphocyte subpopulations in the blood and lymphoid Nawa, Y., 2005. Intraepithelial lymphocytes express junctional
organs of the pig. Immunobiology 201, 7487. molecules in murine small intestine. Biochem. Biophys. Res.
Boes, J., Helwigh, A.B., 2000. Animal models of intestinal nema- Commun. 331, 977983.
tode infections of humans. Parasitology 121 (Suppl.), S97 Kalliomaki, M., Salminen, S., Arvilommi, H., Kero, P., Koskinen, P.,
S111. Isolauri, E., 2001. Probiotics in primary prevention of atopic
Boismenu, R., Havran, W.L., 1994. Modulation of epithelial cell disease: a randomised placebo-controlled trial. Lancet 357,
growth by intraepithelial gamma delta T cells. Science 266, 10761079.
12531255. Kalliomaki, M., Salminen, S., Poussa, T., Arvilommi, H., Isolauri,
Buts, J.P., Bernasconi, P., Vaerman, J.P., Dive, C., 1990. Stimulation E., 2003. Probiotics and prevention of atopic disease: 4-year
of secretory IgA and secretory component of immunoglobulins follow-up of a randomised placebo-controlled trial. Lancet 361,
in small intestine of rats treated with Saccharomyces boulardii. 18691871.
Dig. Dis. Sci. 35, 251256. Kamada, N., Inoue, N., Hisamatsu, T., Okamoto, T., Matsuoka, K.,
Chen, Y., Chou, K., Fuchs, E., Havran, W.L., Boismenu, R., 2002. sato, T., Chinen, H., Su Hong, K., Yamada, T., Suzuki, Y.,
Protection of the intestinal mucosa by intraepithelial gamma Suzuki, T., Watanabe, N., Tsuchimoto, K., Hibi, T., 2005.
delta T cells. Proc. Natl. Acad. Sci. U.S.A. 99, 1433814343. Nonpathogenic Escherichia coli Nissle1917 prevents murine
Christensen, H.R., Frokiaer, H., Pestka, J.J., 2002. Lactobacilli acute and chronic colitis. Inflamm. Bowel. Dis. 11, 455463.
differentially modulate expression of cytokines and maturation Kato, M., Kephart, G.M., Talley, N.J., Wagner, J.M., Sarr, M.G.,
surface markers in murine dendritic cells. J. Immunol. 168, 171 Bonno, M., McGovern, T.W., Gleich, G.J., 1998. Eosinophil
178. infiltration and degranulation in normal human tissue. Anat. Rec.
DSouza, A.L., Rajkumar, C., Cooke, J., Bulpitt, C.J., 2002. Pro- 252, 418425.
biotics in prevention of antibiotic associated diarrhoea: meta- Kruis, W., Fric, P., Pokrotnieks, J., Lukas, M., Fixa, B., Kascak, M.,
analysis. BMJ 324, 1361. Kamm, M.A., Weismueller, J., Beglinger, C., Stolte, M., Wolff,
Gebhardt, T., Sellge, G., Lorentz, A., Raab, R., Manns, M.P., C., Schulze, J., 2004. Maintaining remission of ulcerative colitis
Bischoff, S.C., 2002. Cultured human intestinal mast cells with the probiotic Escherichia coli Nissle 1917 is as effective as
express functional IL-3 receptors and respond to IL-3 by enhan- with standard mesalazine. Gut 53, 16171623.
250 S.C. Duncker et al. / Veterinary Immunology and Immunopathology 111 (2006) 239250

Kruis, W., Schutz, E., Fric, P., Fixa, B., Judmaier, G., Stolte, M., Saalmuller, A., Werner, T., Fachinger, V., 2002. T-helper cells
1997. Double-blind comparison of an oral Escherichia coli from naive to committed. Vet. Immunol. Immunopathol. 87,
preparation and mesalazine in maintaining remission of ulcera- 137145.
tive colitis. Aliment Pharmacol. Ther. 11, 853858. Schultz, M., Strauch, U.G., Linde, H.J., Watzl, S., Obermeier, F.,
Lin, T., Matzusaki, G., Yoshida, H., Kobayashi, K., Kenai, H., Gottl, C., Dunger, N., Grunwald, N., Scholmerich, J., Rath, H.C.,
Omoto, K., Nomoto, K., 1994. CD3-CD8+ intestinal intrae- 2004. Preventive effects of Escherichia coli strain Nissle 1917 on
pithelial lymphocytes (IEL) and the extrathymic development of acute and chronic intestinal inflammation in two different
IEL. Eur. J. Immunol. 24, 10801087. murine models of colitis. Clin. Diagn. Lab. Immunol. 11,
Lodinova-Zadnikova, R., Cukrowska, B., Tlaskalova-Hogenova, H., 372378.
2003. Oral administration of probiotic Escherichia coli after Sinkora, M., Butler, J.E., Holtmeier, W., Sinkorova, J., 2005.
birth reduces frequency of allergies and repeated infections later Lymphocyte development in fetal piglets: facts and surprises.
in life (after 10 and 20 years). Int. Arch. Allergy Immunol. 131, Vet. Immunol. Immunopathol. 108, 177184.
209211. Steidler, L., Neirynck, S., Huyghebaert, N., Snoeck, V., Vermeire,
Loew, D., 2000. Leben und Werken von Alfred Nissle. 1119. A., Goddeeris, B., Cox, E., Remon, J.P., Remaut, E., 2003.
McFarland, L.V., Surawicz, C.M., Greenberg, R.N., Fekety, R., Biological containment of genetically modified Lactococcus
Elmer, G.W., Moyer, K.A., Melcher, S.A., Bowen, K.E., Cox, lactis for intestinal delivery of human interleukin 10. Nat.
J.L., Noorani, Z., 1995. Prevention of beta-lactam-associated Biotechnol. 21, 785789.
diarrhea by Saccharomyces boulardii compared with placebo. Sturm, A., Rilling, K., Baumgart, D.C., Gargas, K., Abou-Ghazale,
Am. J. Gastroenterol. 90, 439448. T., raupach, B., Eckert, J., Schumann, R.R., Enders, C., Sonnen-
Michail, S., Abernathy, F., 2002. Lactobacillus plantarum reduces born, U., Wiedenmann, B., Dignass, A.U., 2005. Escherichia
the in vitro secretory response of intestinal epithelial cells to coli Nissle 1917 distinctively modulates T-cell cycling and
enteropathogenic Escherichia coli infection. J. Pediatr. Gastro- expansion via toll-like receptor 2 signaling. Infect. Immun.
enterol. Nutr. 35, 350355. 73, 14521465.
Miller, E.R., Ullrey, D.E., 1987. The pig as a model for human Vaarala, O., 2003. Immunological effects of probiotics with special
nutrition. Annu. Rev. Nutr. 7, 361382. reference to lactobacilli. Clin. Exp. Allergy 33, 16341640.
Ogra, P.L., Lamm, M.E., Bienenstock, J., Mestecky, J., Strober, W., Vanbelle, M., Teller, E., Focant, M., 1990. Probiotics in animal
McGhee, J.R., 1999. Mukosal Immunology. Academic Press, nutrition: a review. Arch. Tierernahr. 40, 543567.
San Diego. Vega-Lopez, M.A., Arenas-Contreras, G., Bailey, M., Gonzalez-
Pabst, R., Rothkotter, H.J., 1999. Postnatal development of lym- Pozos, S., Stokes, C.R., Ortega, M.G., Mondragon-Flores, R.,
phocyte subsets in different compartments of the small intestine 2001. Development of intraepithelial cells in the porcine small
of piglets. Vet. Immunol. Immunopathol. 72, 167173. intestine. Dev. Immunol. 8, 147158.
Patzer, S.I., Baquero, M.R., Bravo, D., Moreno, F., Hantke, K., 2003. Vega-Lopez, M.A., Telemo, E., Bailey, M., Stevens, K., Stokes,
The colicin G, H and X determinants encode microcins M and C.R., 1993. Immune cell distribution in the small intestine of the
H47, which might utilize the catecholate siderophore receptors pig: immunohistological evidence for an organized compart-
FepA, Cir, Fiu and IroN. Microbiology 149, 25572570. mentalization in the lamina propria. Vet. Immunol. Immuno-
Perdigon, G., Alvarez, S., Rachid, M., Aguero, G., Gobbato, N., pathol. 37, 4960.
1995. Immune system stimulation by probiotics. J. Dairy Sci. 78, von Buenau, R., Jaekel, L., Schubotz, E., Schwarz, S., Stroff, T.,
15971606. Krueger, M., 2005. Escherichia coli strain Nissle 1917: signifi-
Pestka, J.J., Ha, C.L., Warner, R.W., Lee, J.H., Ustunol, Z., 2001. cant reduction of neonatal calf diarrhea. J. Dairy Sci. 88, 317
Effects of ingestion of yogurts containing Bifidobacterium and 323.
Lactobacillus acidophilus on spleen and Peyers patch lympho- Waidmann, M., Bechtold, O., Frick, J.-S., Lehr, H.-A., Schubert, S.,
cyte populations in the mouse. J. Food Prot. 64, 392395. Dobrindt, U., Loffler, J., Bohn, E., Autenrieth, I.B., 2003.
Rembacken, B.J., Snelling, A.M., Hawkey, P.M., Chalmers, D.M., Bacteroides vulgatus protects against escherichia coli-induced
Axon, A.T., 1999. Non-pathogenic Escherichia coli versus colitis in gnotobiotic interleukin-2-deficient mice. Gastroenter-
mesalazine for the treatment of ulcerative colitis: a randomised ology 125, 162177.
trial. Lancet 354, 635639. Westendorf, A.M., Gunzer, F., Deppenmeier, S., Tapadar, D., Hun-
Rodrigues, A.C.P., Cara, D.C., Fretez, S.H.G.G., Cunha, F.Q., ger, J.K., Schmidt, M.A., Buer, J., Bruder, D., 2005. Intestinal
Vieira, E.C., Nicoli, J.R., Vieira, L.Q., 2000. Saccharomyces immunity of Escherichia coli NISSLE 1917: a safe carrier for
boulardii stimulates sIgA production and the phagocytic system therapeutic molecules. FEMS Immunol. Med. Microbiol. 43,
of gnotobiotic mice. J. Appl. Microbiol. 89, 404414. 373384.
Romeis, B., 1989. Mikroskopische Techniken. Urban & Schwarzen- Xu, L.R., Carr, M.M., Bland, A.P., Hall, G.A., 1993. Histochemistry
berg, Munchen. and morphology of porcine mast cells. Histochem. J. 25, 516
Rothkotter, H.J., Ulbrich, H., Pabst, R., 1991. The postnatal devel- 522.
opment of gut lamina propria lymphocytes: number, prolifera- Yang, H., Parkhouse, R.M., 1996. Phenotypic classification of
tion, and T and B cell subsets in conventional and germ-free pigs. porcine lymphocyte subpopulations in blood and lymphoid
Pediatr. Res. 29, 237242. tissues. Immunology 89, 7683.

Das könnte Ihnen auch gefallen