Vous êtes sur la page 1sur 11

IMMUNOLOGY ORIGINAL ARTICLE

Innate profiles of cytokines implicated on oral tolerance correlate


with low- or high-suppression of humoral response

Maria F. Silva,1,2 Alice O. Summary


Kamphorst,3 Elize A. Hayashi,4
Oral tolerance (OT) is being studied with great interest because of its
Maria Bellio,4 Claudia R. Carvalho,3
therapeutic potential in allergy and autoimmunity. In the present study,
Ana M. C. Faria,3 Kátia C. C.
two mouse strains with extreme phenotypes of OT susceptibility (TS) or
Sabino,5 Marsen G. P. Coelho,5
resistance (TR) to ovalbumin (OVA) were used to demonstrate whether
Alberto Nobrega,4 Daniel Tavares5
the tr and ts genes, cumulated during 18 generations of bi-directional
and Antonio C. Silva5
1
genetic selection, influence expression of immunobiological traits in naive
Universidade Estadual do Norte Fluminense,
2 or antigen-gavaged TR/TS mice. The difference in anti-OVA titres was
Universidade Estadual da Zona Oeste, 3Uni-
versidade Federal de Minas Gerais, 4Universid- 2048-fold between OVA-gavaged TS and TR mice. Tolerance susceptibility
ade do Estado do Rio de Janeiro, and to OVA gavage in individuals from a (TS · TR)F2 population was 24%
5
Universidade Federal do Rio de Janeiro, Rio high-susceptibility, 62% low-susceptibility and 14% non-tolerant. Differ-
de Janeiro, Brazil ent antigens, unrelated to OVA, were tested by gavage and TS mice were
generally susceptible while TR mice were resistant. The stability of TS and
TR phenotypes was not affected by the use of strict protocols of intraperi-
toneal immunization or feeding over 30 consecutive days. The levels of
interleukin-2 (IL-2), IL-4, interferon-c and IL-10 cytokines evaluated in
concanavalin A-stimulated spleen cells from naive mice and in OVA-stim-
ulated spleen cells from OVA-gavaged mice were higher in TS mice. Inter-
leukin-10 was up-regulated in OVA-gavaged TS mice and down-regulated
doi:10.1111/j.1365-2567.2010.03248.x in TR mice. In naive mice, the percentage of CD4+ CD25+ and
Received 31 August 2009; revised 15 January CD4+ Foxp3+ spleen cells and IL-10 expression by CD4+ cells was signifi-
2010; accepted 19 January 2010. cantly higher in TS mice. These results indicate that regulation of IL-10
Correspondence: Dr A. C. da Silva,
expression could be an important factor contributing to the mechanisms
Laboratório de Imunobiologia, Department
Biol. Cel. e Genética, Instituto de Biologia,
controlling OT susceptibility, and that the OT responses of TR and TS
UERJ, Rua S. Francisco Xavier, 524, PHLC, individuals strongly correlate with their innate potential to secrete this
Maracanã, CEP 20 559-900, Rio de Janeiro, cytokine.
RJ, Brazil. Email: acdasilva@pq.cnpq.br;
acdasilva.uerj@gmail.com Keywords: antibody responses; cytokines; mice; oral tolerance; regulatory
Senior author: Antonio Carlos da Silva T cells

munity processes and in alleviating allergic inflamma-


Introduction
tory diseases.7,8
Oral tolerance (OT) is a physiological mechanism that The OT regulates responses to established gastrointesti-
prevents food hypersensitivity and adverse allergic nal microflora and continuous antigenic challenges from
responses to normal enteric flora.1,2 Several mechanisms common food antigens.9 Individuals lacking the ability to
have been proposed to explain OT. Low doses of oral induce OT therefore develop immune responses against
antigen favour active suppression with the generation of food antigens and commensal flora and this can trigger
regulatory cells, whereas high doses favour clonal food allergies and inflammatory bowel disease.10 Studies
anergy/deletion3. Regulatory CD4+ T cells producing the in animal models suggest that there is a genetic predispo-
cytokines interleukin-10 (IL-10) or transforming growth sition leading to these diseases.11
factor-b controlling both T helper type 1 (Th1) and In isogenic models of mice, it was demonstrated that the
Th2 responses4,5 mediate specific or non-specific sup- major histocompatibility complex and background genes
pression in antigen-reactive cells.6 The therapeutic control OT.12,13 Our experiments in genetic selection dem-
potential of OT has been tested for systemic autoim- onstrated polygenic control.14 In the present experiment,

 2010 Blackwell Publishing Ltd, Immunology 1


M. F. Silva et al.

we used mice selected for extreme phenotypes of suscepti- F1 hybrids and an F2 segregant population produced by
bility and resistance to OT over 18 generations of consecu- reciprocal crosses between the parental lines TS and TR
tive breeding by assortative mating and avoiding were used in this work. The TS and TR mice were devel-
consanguinity. This strategy favours the selection of coher- oped through bi-directional genetic selection, starting
ent sets of multiple genes participating in the desired OT from a highly polymorphic population (F0) derived from
phenotype. Unlike isogenic strains, in which each mouse the inter-crossing of eight inbred mouse strains (A,
carries a single genome, our tolerance-resistant (TR) and DBA2, P, SWR, CBA, SJL, BALB/c and C57BL/6). Ani-
tolerance-susceptible (TS) mice are genetically homoge- mals used throughout the experiments, and their parents,
neous at the relevant loci for the selected character and het- were maintained on a hen egg OVA-free diet in a produc-
erogeneous in terms of background genes. tion colony, separated from the breeding colony where
An advantage of using animals with extreme pheno- the best phenotypes of susceptibility and resistance to
types is the possibility of reconstitution of an F2 OVA OT were mated for the maintenance of the colony.
(TR · TS) segregant population with a normal phenotype The TR and TS mice were subjected to experimental pro-
distribution in a widely heterogeneous genetic back- tocols and used as parents to produce F1 and F2 mice
ground using inter-strain crosses. Another possibility is after two to three generations of mating in the produc-
the ability to detect phenotypes not found in homoge- tion colony. The Commission for Care and Use of Labo-
neous strains of mice. Multigenic control of the OT phe- ratory Animals of the Rio de Janeiro State University
notype allows different degrees of OT to coexist in a (UERJ, Brazil) approved the study protocols.
heterogeneous population.
In the present study, we analysed the influence of the
Antigens
cumulated tr and ts genes on disparate OT phenotypes in
naive or ovalbumin (OVA) -gavaged TR and TS mice, Ovalbumin and human gamma globulin were purchased
further immunized by intraperitoneal injection with the from Sigma-Aldrich (St Louis, MO). Egg yolk immuno-
ingested antigen, using different protocols for tolerance globulin (IgY) was prepared from eggs inoculated with
induction and immunization. We also studied the genetic canine parvovirus according to McLaren15 while peanut
inheritance of OT trait, analysing the distribution of OT and cashew-nut proteins were extracted as described by
antigen-specific phenotypes in F1 and F2 individuals of a Landry and Moureaux.16 Protein concentrations were
genetically heterogeneous population. The TR and TS determined using the Lowry method.17 A diet containing
mice were tested for tolerance induction by gavage with 15% casein (Rhoster Indústria e Comércio LTDA, SP Bra-
different antigens, unrelated to OVA, measuring their sil) or purified casein (Sigma-Aldrich) was used. Fresh
humoral response after immunization with the respective sheep red blood cells (SRBC) washed in 015 mM NaCl
antigen. The influence of tr and ts genes on the percent- were used immediately. Deaggregated OVA was prepared
age of CD4+ CD25+ and CD4+ Foxp3+ splenic T cells and according to Bruce and Ferguson.18
production of IL-10 by spleen cells from naive and
gavaged TR and TS mice was also evaluated. Other rele-
Tolerance induction and immunization
vant cytokines such as IL-4, interferon-c (IFN-c) and IL-2
were also quantified. The intracellular IL-10 production Mice were gavaged with 02 ml physiological saline con-
by CD4+ T cells from TS mice was found to be greatly taining 5 or 20 mg soluble OVA in a single administra-
augmented compared with TR CD4+ T cells, both in per- tion 7 days before immunization. Control groups were
centages and in levels of secretion. Percentages of regula- gavaged with saline alone. Continuous feeding of 20 mg
tory T (Treg) cells, measured by CD25 and Foxp3 OVA in the drinking water was given for 1, 3 or 30 con-
markers, were higher in TS mice. These results clearly secutive days (each mouse ingested 20 mg OVA daily).19
indicate that the cumulated tr and ts genes control the Seven days after the last day of oral treatment, mice
innate profile of cytokine production and Treg cell per- were immunized intraperitoneally (i.p.) with 10 lg
centages of naive TS and TR animals, before oral treat- OVA + 1 mg alum and 14 days after with one booster of
ment with OVA, and strongly correlate with OT 10 lg OVA without alum. To evaluate the effect of con-
susceptibility or resistance obtained after being fed and secutive i.p. immunization on OT maintenance, mice ga-
further immunized with antigen. vaged with OVA 5 mg and i.p. immunized 7 days after
with 10 lg OVA + 1 mg alum were boosted with i.p.
10 lg soluble OVA on days 14, 21 and 28 after the first
Materials and methods
immunization. Sera were collected 7 days after the last
immunization. To induce endovenous tolerance, 02 ml
Mice
physiological saline containing 2 mg deaggregated OVA
Two- to three-month-old mice of both sexes from an F18 was injected in a single intravenous (i.v.) dose 7 days
generation of OT-susceptible (TS) or OT-resistant (TR) before i.p. immunization. To induce oral tolerance to

2  2010 Blackwell Publishing Ltd, Immunology


Naive mice and OVA oral tolerance

SRBC, we followed the protocol of Mowat.20 Sera were was developed at room temperature with 100 ll/well of
collected 21 days after immunization. orthophenylenediamine (1 mg/ml), 004% H2O2 substrate
in sodium citrate buffer. Reaction was interrupted by the
addition of 20 ll/well 2 M H2SO4. Absorbance was mea-
Evaluation of humoral response
sured at 490 nm using an ELISA reader (Bio-Rad Model
The OVA-specific agglutinin titres of F2 populations were 450 Microplate Reader, Hercules, CA).
determined by passive haemagglutination with SRBC-cou-
pled OVA according to Avrameas et al.21 The SRBC-spe-
Cell staining and flow cytometry
cific agglutinin titres of TR and TS mice were determined
by direct haemagglutination testing. The titres were Freshly isolated spleen cells obtained on the third day
expressed as log2 of the highest serum dilution giving a after immunization from naive or previously saline-
positive reaction. Immunoglobulin G antibodies of TR gavaged or OVA-gavaged TR and TS mice were stained
and TS mice to OVA and other antigens such as casein with fluorescein isothiocyanate-labelled anti-mouse CD4
(Sigma-Aldrich), fowl gamma globulin (IgY), peanuts and (clone RM4-5; Pharmingen) and phycoerythrin-Cy5-
cashew nuts were assessed by enzyme-linked immunosor- labelled or allophycocyanin-labelled anti-mouse CD25
bent assay (ELISA) as described previously.22 Tetrameth- (clone PC61; eBioscience, San Diego, CA) for analysis, or
ylbenzidine (Zymed Lab., Inc., San Francisco, CA) was with phycoerythrin-Cy7-labelled anti-CD19 (clone
used as the substrate for horseradish peroxidase. The yel- MB19.1; eBioscience) and Alexa 647 IgM (clone b-7-6;
low colour produced was read at 450-nm wavelength kindly provided by Dr John Cambier, University of Colo-
using a multiscan photometer. Experimental and control rado Health Science Center, Denver, CO).
groups were evaluated by comparing the mean ± SD of Typically, 1 · 106 to 3 · 106 erythrocyte-depleted
the sums of absorbance values read between 1/800 and 1/ splenocytes in suspension were incubated in fluorescence-
25 600 serum dilutions (the most linear part of the absor- activated cell sorting (FACS) buffer (PBS, 1% FCS, 005%
bance curve).22 The mean values were therefore based on sodium azide) with mAbs in appropriate dilutions for
readings of six dilutions of each individual serum. 20 min on ice, and then washed with FACS buffer. For
FoxP3 staining, cells were further fixed and permeabilized
using the mouse regulatory T-cell staining kit (eBio-
Cell proliferation and cytokine assays
science), following the manufacturer’s instructions. Data
Spleen cells from individual mice were harvested 14 days were acquired on a FACSCalibur (BD Biosciences, San
after immunization and cultured in 96-well plates at Jose, CA) and analysed using CELLQUEST (BD Bioscienc-
106 cells/ml in the presence of anti-CD3 and anti-CD28 es) or SUMMIT (Dako Cytomation, Glostrup, Denmark)
antibodies (1 lg/ml each), concanavalin (Con A) (1 lg/ software.
ml), OVA 01, 1 or 5 lg/ml for 72 hr, or Escherichia coli
lipopolysaccharide 125, 25, 5 or 10 lg/ml (LPS; Sigma-
Con A blast generation and intracellular staining (ICS)
Aldrich). [3H]Thymidine (1 lCi/well) was added for the
for IL-10 production
last 6 hr of culture and the incorporated thymidine was
measured in a beta liquid scintillation counter. For cyto- Splenocytes from TR and TS mice were cultured for
kine assays, culture supernatants were collected at 24 hr 2 days in complete medium (OptiMEM medium contain-
for IL-2 and at 72 hr for IFN-c, IL-4 and IL-10. Levels of ing 10% FCS, 100 lg/ml penicillin, 100 U/ml streptomy-
cytokines in supernatants were determined by capture cin, and 50 nM 2b-mercaptoethanol (all from Life
ELISA as described elsewhere.23 Briefly, 96-well microtitre Technologies Inc., Grand Island, NY) in the presence of
plates (NUNC, Naperville, IL), were coated with 100 ll/ Con A (25 lg/ml; Sigma-Aldrich), followed by 3 days of
well of respective capture antibodies (Pharmingen, San culture in rIL-2 (20 ng/ml) and rIL-4 (20 ng/ml) (R&D
Diego, CA) at a dilution of 2 lg/ml in coating buffer, and Systems, Inc., Minneapolis, MN) to generate Con A
incubated overnight at 4. The plates were then washed blasts. For IL-10 detection, Con A blasts obtained as
with phosphate-buffered saline (PBS) –01% Tween-20 described above were further stimulated with phorbol 12-
and blocked for 60 min at 37 with 10% fetal calf serum myristate 13-acetate (PMA; 20 ng/ml), calcium ionophore
(FCS). Culture supernatants and standards were loaded A 23187 (200 ng/ml) and monensin (5 lM) for 5 hr. Cells
onto plates. The plates were thoroughly washed and the were then pre-stained for cell surface markers, followed
appropriate biotinylated rat anti-mouse IL-2, IL-4, IL-10 by fixation in 1% paraformaldehyde for 18 hr at 4 and
or IFN-c monoclonal antibodies (mAbs; Pharmingen) permeabilization with 02% saponin in FACS buffer
were added, followed by a 60-min incubation at 37. After (PBS-supplemented with 5% FCS and 005% sodium
additional washes, horseradish peroxidase-labelled strepta- azide) for 20 min at 4. Cells were then incubated for
vidin (Sigma-Aldrich) was added to each well for 15 min 20 min with phycoerythrin-conjugated anti-mouse IL-10
and each plate was thoroughly washed. Colour reaction mAb or isotype control (eBiosciences) properly diluted in

 2010 Blackwell Publishing Ltd, Immunology 3


M. F. Silva et al.

02% saponin FACS buffer, on ice. Alternatively, freshly 20 mg OVA for 30 days (Fig. 1b). The TR mice showed a
isolated splenocytes from TR and TS mice were directly steady resistance after any of these different regimens
resuspended (25 · 106 cells/ml) in complete medium whereas TS mice became tolerant on all feeding regimens.
with LPS (10 lg/ml, E. coli serotype 0111:B4), PMA Continuous feeding for 1 or 3 days was more effective
(50 ng/ml), calcium ionophore A 23187 (200 ng/ml) and than gavage to down-regulate the in vivo IgG response.
monensin (5 lM) (all from Sigma-Aldrich) for 5 hr, in
24-well flat-bottom plates for detection of IL-10 produc-
The stability of TS and TR mouse phenotypes was
tion by splenic B lymphocytes, as previously described.24
not changed after continuous i.p. challenge with
Cells were pre-stained for B-cell surface markers and pro-
OVA + alum or OVA + complete Freund’s adjuvant
cessed as above for intracellular IL-10 staining.
To test the robustness of the selection, the TS and TR
mice were evaluated under immunogenically extreme
Genetic analysis
conditions. Figure 1(c) shows the kinetics of primary and
The degree of dominance effect in the F1 (TR · TS) secondary responses of previously OVA-gavaged mice.
hybrid was calculated as d/a. The d (dominance devia- The secondary i.p. challenge did not induce a humoral
tion) represents the distance from the mid-parental value response in OVA-gavaged TS mice. The OVA-gavaged or
caused by dominance and is calculated according the saline-gavaged TR mice and the saline-gavaged TS mice
equation: d = xF1 ) ½(xA + xB), where xA and xB are responded to the secondary challenge with augmented
the mean response of each parent. The determination of specific IgGs. To confirm the stability of susceptibility to
a (additive effect) might be obtained by measuring OVA OT, TS and TR mice gavaged with 5 mg OVA were
the departure from the F1 hybrid mean to either of boosted by i.p. injections of 10 lg OVA in saline on days
the parents, or halving the parental difference: a = ½ 14, 21 and 28 after the first i.p. immunization with 10 lg
(xA ) xB).25 The F1 mean value would be expected to be OVA + complete Freund’s adjuvant (CFA; Fig. 1d). The
exactly intermediate to both parents if the genetic effects IgG response of OVA-gavaged TS mice stayed stable
were additive, departures from this mid-parental value throughout despite the rigorous immunization protocol.
would indicate the effect of dominance. The value There was no significant difference in antibody levels
between zero and one calculated by the ratio d/a signifies between OVA-gavaged and saline-gavaged TR mice.
the dominance effect.
TS mice were generally susceptible and TR mice were
Statistical analysis resistant to tolerance induction for the majority of
non-related antigens tested
The chi-square goodness-of-fit test was used to evaluate
whether the observed data were coherent with a normal To test the responses to different routes of sensitization,
curve. Differences between mean antibody levels in differ- or to antigens not used during the selection process, TR
ent mouse groups were determined by Student’s t-test; and TS mice were gavaged using different and unrelated
groups were considered statistically different if P  005. soluble [OVA, bovine serum albumin (BSA), IgY, human
gamma globulin, casein, peanut and cashew-nut extracts]
and particulate (SRBC) antigens (Table 1); intravenous
Results
injection with OVA was also evaluated. Following these
treatments, mice were immunized with the homologous
Feeding OVA for 30 consecutive days did not inhibit
antigens and the sera were obtained to measure levels of
the IgG response in TR mice
IgG antibodies as described in the Materials and methods.
In the present study, the tolerogenic potential of mice The results were as follows. (i) The IgG response differ-
selected for extreme phenotypes of oral tolerance (TR and ences, between gavaged TS and TR mice (inter-strain dif-
TS mice) was evaluated. The mean of specific IgG titres ference), were statistically significant to all tested antigens
to antigen (OVA) used for selecting these strains of mice except to BSA. (ii) Differences in specific IgG responses
was significantly different. In the present generation, the between antigen-gavaged and saline-gavaged TS mice, fol-
difference was 2048-fold between OVA-gavaged TS and lowed by immunization with the same antigen used to
TR mice and 32-fold between saline-gavaged TS and TR gavage, were statistically significant for casein, peanut,
mice. BSA, SRBC, IgY and OVA. Cashew nuts and human
We evaluated the phenotypic profiles of TS and TR gamma globulin did not induce OT. The TR mice did
mice when submitted to different oral treatment proto- not show differences in antibody titres between experi-
cols: each mouse received a single dose of 5 mg OVA by mental and control groups excluding BSA. (iii) Intrave-
gavage, or daily 5 mg OVA diluted in drinking water nous OVA only induced tolerance in the TS mice
(continuous feeding) in 1 or 3 days (Fig. 1a) or daily (Table 1).

4  2010 Blackwell Publishing Ltd, Immunology


Naive mice and OVA oral tolerance

(a) (b)
9 TS mice 16
TR mice 14
6

lgG titres (log2)

lgG titres (log2)


*** *** 12
3 *** 10
0
8
9
6 **
6 4
3 2
0 0
Gavaged Fed OVA fed 30 days
1d 3d
OVA – + + + – + – +

(c) (d)
20 Primary Secondary
18 10
TS mice
lgG titres (log2)

16

lgG titres (log2)


TR mice 8
14
6
12
10 4
8 2
6
0
14 21 28 14 21 28 14 21 28 35
Days after i.p. immunization Days after i.p. immunization

Figure 1. Consecutive oral tolerance induction and intraperitoneal immunization. (a) Mice were gavaged with a single dose of ovalbumin (OVA;
5 mg), or submitted to continuous OVA feeding for 1 or 3 consecutive days. (b) Mice received 20 mg OVA by continuous feeding daily for 30
consecutive days. In (a) and (b) mice were intraperitoneally (i.p.) immunized with 10 lg OVA + 1 mg alum 7 days after OVA oral treatment.
(c) Mice were gavaged with 5 mg OVA and given a single i.p. immunization with 10 lg OVA + 1 mg alum (primary response) or were i.p.
immunized and boosted with 10 lg OVA (secondary response). (d) Mice were gavaged with 5 mg OVA and i.p. immunized with 10 lg
OVA + CFA and boosted three times, at 7-day intervals, with 10 lg OVA. Arrows indicate booster days. Squares indicate OVA-fed group and
the circles indicate saline-fed group. Results represent mean ± SD of two experiments with eight mice per group. The differences in antibody
titres between OVA-fed TS mice and the other groups were statistically significant at all time-points (P < 001 by Student’s t test).

Table 1. Humoral response to unrelated antigens

Log2 immunoglobulin G titres – intragastric pre-treatment

Tolerance-susceptible (TS) mice Tolerance-resistant (TR) mice

Antigens Saline1 Antigen2 P-value3 Saline4 Antigen5 P-value6 P-value7

Ovalbumin (OVA) 1230 ± 130 600 ± 147 P < 0001 1470 ± 150 1310 ± 270 NS P < 0001
Casein 826 ± 287 418 ± 329 P < 0005 823 ± 285 903 ± 129 NS P < 0001
Peanuts 1150 ± 220 360 ± 180 P < 0001 1320 ± 250 1230 ± 200 NS P < 0001
Cashew nuts 900 ± 120 810 ± 230 NS 1240 ± 220 1200 ± 210 NS P < 0001
Sheep red blood cells (SRBC) 1100 ± 141 800 ± 219 P < 0001 1200 ± 100 1260 ± 151 NS P < 0001
Fowl gamma globulin (IgY) 875 ± 075 770 ± 030 P < 0001 1080 ± 060 1122 ± 017 NS P < 0001
Human gamma globulin (HGG) 870 ± 114 848 ± 123 NS 1117 ± 118 1282 ± 090 NS P < 0001
Bovine serum albumin (BSA) 954 ± 093 743 ± 321 P < 005 857 ± 315 567 ± 296 P < 005 NS
Log2 IgG titres – intravenous pre-treatment
Deaggregated OVA 1156 ± 136 482 ± 219 P < 0001 1300 ± 040 1288 ± 037 NS P < 0001

NS, not significant difference, P > 0.05. Mean value with standard deviation.
1
TS mice saline-gavaged.
2
TS mice antigen-gavaged.
3
Differences between antigen-gavaged and saline-gavaged TS mice.
4
TR mice saline-gavaged.
5
TR mice antigen-gavaged.
6
Differences between antigen-gavaged and saline-gavaged TR mice.
7
Inter-strain differences between antigen-gavaged TR and TS mice.
Results represent means ± SD of 10 mice per group.

 2010 Blackwell Publishing Ltd, Immunology 5


M. F. Silva et al.

(a) (b) (c) (d)


35 1·0 8
30 TS mice 7 0·3

IFN-γ (ng/ml)
c.p.m. (× 103)
TR mice

IL-10 (ng/ml)
0·8 6

IL-2 (ng/ml)
25
20 0·6 5 0·2
15 * ** 4
0·4 3
10 2 0·1
5 0·2
1
0·0 0 0·0
35
30 1·0 8
c.p.m. (× 103)

25 7 0·3

IFN-γ (ng/ml)
0·8

IL-10 (ng/ml)
6

IL-2 (ng/ml)
20
0·6 5
15 4 0·2
** **
10 0·4 ** 3
0·1
5 2
0·2 **
0 1
i ii iii i ii iii i ii iii 0·0 0 0·0
Saline Gavage Feeding Saline Gavage Feeding Saline Gavage Feeding Saline Gavage Feeding

Figure 2. Specific proliferative responses and cytokine production. Mice received 5 mg ovalbumin (OVA) by either gavage or continuous feeding
for 24 hr and were immunized subcutaneously with 10 lg OVA + complete Freund’s adjuvant (CFA) 7 days later. Control mice received oral sal-
ine and were immunized. Spleen cells were collected 14 days after immunization and 106 cells/well were cultured in 96-well plates in the presence
of anti-CD3 and anti-CD28 antibodies in medium containing 01, 1 or 5 lg/ml OVA for measuring the proliferative response (a column i, a col-
umn ii and a column iii, respectively) and 1 lg/ml OVA for cytokine production (b–d). Bars represent the mean ± SD of three experiments with
eight mice per group. *P < 005, **P < 001 comparing experimental with control groups.

Oral treatment with OVA affects the in vitro specific T-cell and B-cell proliferative responses and cytokine
proliferation of spleen cells from TS and TR mice production differed in naive TS and TR mice
The specific proliferative response of spleen cells from TS To rule out the possibility that TS mice were simply low
mice submitted to gavage or continuous feeding (Fig. 2a) responders, the polyclonal proliferative response and cyto-
was inhibited significantly when compared with saline- kine production were studied in naive TS and TR mice
gavaged TS mice. Although OT was more effective in TS after in vitro stimulation of spleen cells. To induce B-cell
mice, suppression of specific proliferation was induced in cell proliferation splenocytes were stimulated in medium con-
cultures from OVA-gavaged TR mice stimulated with taining LPS. B-cell proliferation was lower in TS mice
01 lg/ml OVA (Fig. 2a, column i). The cells from TR mice when 125, 25 or 5 lg/ml LPS were used (P < 0001)
submitted to OVA continuous feeding showed increased (Fig. 3a). To investigate T-cell proliferation and cytokine
proliferation when stimulated with 1 lg/ml OVA if com- production, splenocytes were cultured in medium con-
pared with the saline-gavaged TR group, suggesting oral taining Con A or anti-CD3/CD28 (Fig. 3b–f). T-cell pro-
priming (Fig. 2a,b). Proliferation of spleen cells from saline- liferation was higher in TS mice when anti-CD3/CD28
gavaged mice was higher in TS than TR mice, after 1 or 5 lg/ stimulated (P = 0002) (Fig. 3ab). The production of
ml OVA stimulation (Fig. 2a, columns ii, iii). However, IL-2, IFN-c, IL-4 and IL-10 cytokines by T cells were
OVA 01 lg/ml did not stimulate TS spleen cells (Fig. 2a). significantly higher in TS than TR mice (Fig. 3c–f).

IL-2, IL-10 and IFN-c levels were reduced in OVA- Inter-strain differences in CD25+ and Foxp3+ CD4+
gavaged TR mice T-cell frequency
We investigated the effect of OVA administration by either Naive or saline-gavaged TR mice showed a higher CD4+
gavage or continuous feeding on cytokine secretion by T-cell count (Fig. 4a) but a lower relative CD25+ percent-
spleen cells from TR and TS mice. Gavage led to decreased age than naive or saline-gavaged TS mice (Fig. 4b). These
IL-2 production in both strains of mice (Fig. 2b). High lev- inter-strain differences disappeared after OVA-gavage as
els of IFN-c and IL-10 were observed in TS mice result of a reduction in CD4+ cells in TR mice (Fig. 4b,c).
(Fig. 2c,d). On the other hand, IFN-c and IL-10 levels were Despite differences in the proportion of CD25+ within
reduced in OVA-gavaged TR mice (Fig. 2c,d). Interleukin- CD4+ T cells, no significant difference between TR and
10 was also reduced in OVA-fed TR mice (Fig. 2D). Reduc- TS mice was observed in the proportion of CD25+ cells
tion in IFN-c and IL-10 production (Fig. 2c,d) may be among total splenocytes (Fig. 4c). Similar to our observa-
mediating the TR mouse resistance to OT, as observed by tions with CD25, we also found that the Foxp3+ cell pro-
increased proliferation of spleen cells harvested from OVA- portion within the CD4+ T-cell population from naive TS
fed TR mice (Fig. 2a, column ii). mice was significantly higher than in TR mice (Fig. 4d).

6  2010 Blackwell Publishing Ltd, Immunology


Naive mice and OVA oral tolerance

(a) 6 (b) (c) 8


TR mice 160
6
4 TS mice 120
4
80
2 *** *** *** 40
2
c.p.m. (× 103)

c.p.m. (× 103)

IL-2 (ng/ml)
0 0 0

160 8
6
120 6
4
80 4
***
2
40
** 2 **
0 0 0
0 1·25 2·5 5 10 Con A α-CD3/CD28 Con A α-CD3/CD28
LPS concentration (µgml)

(f)
(d) (e)
20
30 0·4
16

20 12
8 0·2
10
IFN-γ (ng/ml)

4
IL-10 (ng/ml)

IL-4 (ng/ml)
0 0 0·0
20
30 0·4
16

20 12
8 * 0·2
10 *** *** 4 ***
**
0 0 0·0
Con A α-CD3/CD28 Con A α-CD3/CD28 Con A

Figure 3. Proliferative response to concanavalin A (Con A), anti-CD3/CD28 antibody and lipopolysaccharide (LPS), and cytokine production
from naive mice. For proliferative response, 106 spleen cells/well were cultured (a) in medium containing LPS 125, 25, 5 or 10 lg/ml, or (b–f)
in medium containing anti-CD3/CD28 antibody or Con A. (c–e) interleukin-2 (IL-2), interferon-c (IFN-c) and IL-10 were stimulated by Con A
or anti-CD3/CD28 antibody. (f) IL-4 was stimulated by Con A. Bars represent the mean ± SD of three experiments with six mice per group.
*P < 005, **P < 001, ***P < 0001 comparing experimental tolerance-susceptible (TS) and tolerance-resistant (TR) groups.

(a) (b) (c) (d)


CD25+ in the spleen (%)

40 P = 0·03
CD25+ T cells (%)
CD4+ T cells (%)

P < 0·01
CD4+ Foxp3+ (%)

P < 0·01 50 P < 0·01 P < 0·01 14


8
30 40 12
6
30 10
20 4
20 8
2 6
10 10
TR TS TR TS TR TS TR TS TR TS TR TS TR TS TR TS TR TS TR TS
Naive Saline OVA Naive Saline OVA Naive Saline OVA Naive

Figure 4. Frequency distribution of CD4+, CD25+ and Foxp3+ T cells measured by flow cytometry. Three experimental groups are shown: naive
(non-immunized) and saline or ovalbumin (OVA) -gavaged [both intraperitoneally (i.p.) OVA-immunized]; (a) CD4+ cells as a percentage of
gated splenic lymphocytes, (b) CD25+ cells as a percentage of gated CD4+ cells. (c) CD25+ cells as a percentage of spleen cells. (d) CD4+ Foxp3+
T-cell frequency. A representative evaluation with six mice of each group is shown.

(CD19+ IL-10+) (Fig. 5a). However, a greater proportion


Higher IL-10 levels were produced by CD4+ T cells
of IL-10+ non-B cells was detected in TS mouse spleno-
from TS mice
cytes (Fig. 5b). We aimed to evaluate whether such an
No difference between naive TS and TR mice was inter-strain difference was the result of an intrinsic defect
observed in the proportion of IL-10-producing B cells of CD4+ T cells in the response to IL-10-inducing stimuli,

 2010 Blackwell Publishing Ltd, Immunology 7


M. F. Silva et al.

(a) 6 (b) 4
P = 0·004

% IL-10 (non-B cells)


% IL-10 (B cells)

3
4

3
2
2

1 1 Figure 5. Frequency distribution of CD4+ IL-


TR TS TR TS 10+ and CD19+ IL-10+ cells and interleukin-10
(IL-10) mean fluorescence intensity (MFI)
(c) P < 0·0001 P < 0·0001 from tolerance-susceptible (TS) and tolerance-
(d)
20 100 resistant (TR) spleen cells analysed by flow
cytometry. (a,b) Interline comparative propor-
MFI CD4+ IL-10+ (%)

tion of CD19+ IL-10+ and CD19- IL-10+ cells


CD4+ IL-10+ (%)

15
in freshly isolated spleen between naive TS and
80 TR mice. (c,d) Inter-strain difference between
10 TS and TR mice on proportion of CD4+ IL-
10+ T cells and intracellular IL-10 levels by
5 CD4+ T cells after in vitro stimulation for gen-
60 eration of IL-10-producing blasts. A represen-
0 tative evaluation with six mice of each group is
TR TS CtrTR CtrTS TR TS shown.

using an in vitro treatment that favours the generation of males are, respectively, used for mating. No significant
IL-10-producing blasts from splenic CD4+ T cells. A difference was observed between OVA-gavaged and sal-
higher proportion of CD4+ IL-10+ T cells and intracellu- ine-gavaged F1 mice.
lar IL-10 production by CD4+ T cells were observed in The difference between OVA-gavaged and saline-
naive TS mice (Fig. 5c,d). gavaged F2 populations (Fig. 6d,h) was highly significant.
To differentiate tolerogenic or non-tolerogenic mice in an
OVA-gavaged F2 population we used one standard devia-
Genetic inheritance of OT susceptibility in TR, TS, F1
tion of the feeding TR and TS means as values to estab-
and F2 mice and the dominance effect observed in F1
lish these distinct groups. The high-tolerance individuals
hybrids gavaged with OVA
had values lower than 80 log2 (24%), the high-resistance
TS and TR mice, and their F1 and F2 populations result- mice had values over 130 log2 (14%) and the low-toler-
ing from interbreeding TR · TS, underwent tolerance ance (62%) comprised a normal F2 population (Fig. 6d).
induction and immunization protocols, and the titres of The IgG titres were distributed according to a Gauss nor-
serum IgG anti-OVA obtained were evaluated. Compari- mal distribution.
sons of the frequency distributions of IgG titres between The genetic significance of the overlap between the
TR and TS OVA-gavaged mice and with their respective phenotypic frequencies of F1 and TR (Fig. 6b,c) was esti-
saline-gavaged control groups, and between OVA-gavaged mated by calculating the degree of dominance effect using
F1 and F2 populations with their saline-gavaged control the mean of the responses of TS, TR and F1 hybrids trea-
groups, are shown in Fig. 6. We observed a left bias in ted by gavage with OVA. Using the equation d = xF1 ) ½
the gavaged TS IgG titre distribution, suggesting a critical (xA + xB), as described in Material and methods, demon-
value for maximal OT induction in the population of strates an additive effect of a = 5015, a dominance devia-
nearly 5 log2 (Fig. 6a). tion of d = 2725, and a dominance effect of d/a = 054.
To evaluate if maternal effects influenced the immuno- This value ensures the dominance effect of OT-resistance
logical expression of naive or antigen-stimulated TR and over the OT-susceptible phenotype measured by IgG level
TS mice, the F1 hybrids produced by reciprocal crosses (Fig. 6b,c).
were gavaged with OVA or saline. No significant differ-
ence (P = 050) was observed between the means of F1
Discussion
offspring from TR or TS mothers (1454 ± 166 and
1420 ± 152, respectively), indicating non-maternal influ- Models of quantitative variation in immune responses
ence. Therefore, offspring of TS females can express have explained phenotypic attributes successfully for
OT-resistant or OT-susceptible phenotypes if TR or TS many biological systems25 and OT phenotypes can be

8  2010 Blackwell Publishing Ltd, Immunology


Naive mice and OVA oral tolerance

60 TS mice
(a) 30 TS mice (e)
40 20
20 10
0 0
5 10 15 20 5 10 15 20
60 TR mice

Frequency distribution
(b) 30 (f)

Frequency distribution
40 TR mice
20
20 10
0 0
5 10 15 20 5 10 15 20
10 (c) 8 (g)
F1 mice 6 F1 mice
5 4
2
0 0
5 10 15 20 5 10 15 20
20 F 2 mice L = 62% (d) 20 (h)
15 15 F2 mice
10 S = 24% R = 14% 10
5 5
0 0
5 10 15 20 5 10 15 20
Agglutinin titres (Log2) Agglutinin titres (Log2)
OVA-gavaged mice Saline-gavaged mice

Figure 6. Frequency distribution of agglutinin titres from tolerance-susceptible (TS) and tolerance-resistant (TR) mice, F1 hybrid and F2 popula-
tion. Mice were previously ovalbumin (OVA) -gavaged (a–d) or saline-gavaged (e–h) and presented the following immunoglobulin G (IgG) titres
mean ± SD. (a) OVA-gavaged TS = 662 ± 187 (81 mice); (b) OVA-gavaged TR = 1665 ± 228 (102 mice); (e) saline-gavaged
TS = 1042 ± 174 (52 mice); (f) saline-gavaged TR = 1613 ± 165 (66 mice). In F1 populations, the values were: (c) OVA gavaged
F1 = 1436 ± 157 (28 mice) and (g) saline-gavaged F1 = 1419 ± 204 (18 mice). In F2 populations, the values were: (d) OVA-gavaged
F2 = 1008 ± 249 (89 mice) and (h) saline-gavaged F2 = 1142 ± 204 (47 mice). The mean differences between OVA-gavaged TS and OVA-
gavaged TR were statistically significant (P < 0001) by t-test as were those between OVA-gavaged TS and its respective saline-gavaged control
(P < 0001), and between OVA-gavaged F2 and saline-gavaged F2 (P < 001). There was no significant difference between OVA-gavaged and sal-
ine-gavaged F1 or between F2 groups so as between OVA-gavaged F1 and OVA-gavaged TR mice. H, high susceptible; L, low susceptible; R, resis-
tant.

understood through them. The goal of a trait-related for the induction of OT.27 None of these regimens was
genetic selection is to create mouse strains with extreme able to abrogate resistance to OT in the TR mice. This
and easily differentiated phenotypes. Although we selected indicates that TR mice are strongly resistant, and TS mice
TS and TR mice in response to feeding with OVA, it is are susceptible, not only to protocols used during the
clear from results that susceptibility or resistance to OT selective process but to others that are apparently more
are not restricted to OVA, but is a general property of efficacious (Fig. 1; Table 1).
the TS and TR strains (Table 1). Tolerance susceptibility Because the physiology of antigen absorption and pre-
for multiple immunogens suggests that the outcome of sentation in the intestinal mucosa is peculiar, we investi-
the genetic selection based on a single antigen could not gated whether TS and TR mice would maintain their
be dissociated from a more profound alteration of the tolerance phenotypes for intravenous OVA. The suscepti-
immune response. ble and resistant phenotypes were unchanged (Table 1).
To investigate if OT susceptibility could be maintained Therefore, oral and intravenous tolerance could share
by using different regimens of immunization, TS mice fundamental mechanisms, involving organs such as the
were treated with sequential immunogenic boosters with liver and processing and presentation by specialized
OVA (Fig. 1c,d). In these immunization protocols, we cells.28
used alum or CFA as adjuvant in accordance with the Oral tolerance is a mechanism that probably arose to
observation by Tobagus et al.26 that the nature of inflam- protect the organism against inflammatory diseases such
matory co-stimulation by adjuvant used to express spe- as allergies and autoimmune disorders,2,7,29 so absolute
cific responses after feeding antigens determines the resistance to OT would not be compatible with life
qualitative aspects of the tolerance process. The tolerogen- under normal conditions and we are unlikely to find
ic capacity of TS mice was not modified by boosters or animals resistant for all immunological parameters in
by use of different adjuvant. The TR mice treated either natural populations. We observed that in special condi-
by gavage with low or high doses of OVA, or submitted tions the TR mice were primed or suppressed depend-
to continuous feeding for short and long periods were ing on whether high or low doses of OVA were given
not tolerized (Fig. 1a,b). Continuous feeding resembles orally. We found both down-regulation and up-regula-
natural feeding and is more effective than gavage to sup- tion in antigen-specific cellular proliferation (Fig. 3a), as
press antibody production. Continuous uptake of small in previous results for delayed-type hypersensitivity reac-
amounts of antigen in physiological conditions can signal tion and IgE responses.22,29 This dose-dependent

 2010 Blackwell Publishing Ltd, Immunology 9


M. F. Silva et al.

suppressor or enhancer capacity of TR mice was also moelcule, IL-1 and IL-12 levels and acute inflammatory
observed as bystander OT for Leishmania amazonensis responses39, probably as a consequence of low IL-10 lev-
infection.30 els, and these molecules can signal resistance to suppres-
The cytokines IFN-c, IL-4, IL-10 and transforming sion.
growth factor-b have been implicated as active mediators The quantitative character of OT was demonstrated by
of oral tolerance. They are associated with the develop- the normal curve of the heterogeneous population used
ment of Th1, Th2 or Th3 responses and generation of to start the selection, and by successful production of
regulatory T cells.31–33 The OVA-gavaged and OVA-fed mice with extreme phenotypes of OT susceptibility and
TS mice maintained high levels of IL-10, IL-4 and IFN-c resistance (TS and TR strains) selected during 18 genera-
and OVA-gavaged TR mice showed reduced IFN-c and tions of assortative mating (Fig. 5a–c,e).14 The reconstitu-
IL-10 secretion levels concomitant with the decreased tion of a segregant F2 population with a normal
IL-2 levels (Fig. 2b,d). The TS naive mice also produced phenotype distribution in a widely heterogeneous genetic
higher in vitro IL-4, IL-10 and IFN-c levels as along with background and its use in OT studies allows determina-
T-cell proliferation than did TR mice (Fig. 3b–f), but tion of the frequency of well-defined phenotypic groups
lower B-cell proliferation (Fig. 3a) in response to LPS. and can be useful for therapeutic studies in human popu-
We found that TS mice produce IL-10 after intravenous lations. (Fig. 5d,h). As TR and TS phenotypes were
LPS (unpublished work). Results of other authors show selected to OVA-specific OT, we produced F1 hybrids by
the relationship of OT and bacterial products of intestinal reciprocal crosses to test if maternal antibodies could
flora and that LPS administered at the time of OVA interfere on the immunological expressions of naive or
ingestion increases OT.34 Indeed, these responses in antigen-stimulated mice. Both TR and TS mothers pro-
naive animals may represent the background response duced OT-resistant offspring (Fig. 5c,g) in these crosses.
to environmental antigens that come into contact with The OT-resistant offspring from mating TS females with
the immune system via the intestinal and respiratory TR males, and OT-susceptible offspring from TS females
mucosas. with TS males, therefore indicate no maternal interference
Naive CD4+ T cells can differentiate into either IFN-c- in the studied mice.
producing Th1 cells or IL-4-producing Th2 cells, and that The F1 hybrid response closer to that of the TR mice
reciprocal regulation of Th1 and Th2 responses is medi- (dominance effect), suggests epistatic deviation of OT
ated by IL-4 and IFN-c,35 both of which antagonize Th17 genes in the direction of the TR mouse phenotype
differentiation.36 This subset of CD4+ Th cells, called (Fig. 5a–c). Recent results show that the TS mice are low-
Th17, was implicated in the protection of the host against inflammatory mice while TR and the F1 mice are fivefold
extracellular pathogens encountered at mucosal surfaces, more inflammatory than TS mice39. This is suggestive of
and plays a detrimental role in autoimmune disorders, as an association between OT and inflammation. Hence, the
well as in human inflammatory bowel disease and psoria- opposite capacity of the genetically modified mice may be
sis.37 The information that IL-4 and IL-10 inhibit the Th1 involved in co-adaptive mechanisms, reflecting a dynamic
responses, and that IL-4, IL-10 and IFN-c are required to relation between gene frequencies in a natural popula-
control the Th17 responses,38 suggest the participation of tion.29
these cytokines in establishing phenotypes of specific OT Our results reinforce the importance of regulatory T
susceptibility/resistance and in the diverse inflammatory cells producing IL-10 as key players in OT. We suggest
responses of TR and TS mice.39 that the disparate immunological profiles of naive or fed
Regulatory T lymphocytes (Treg cells) are important TS and TR mice are the result of OT-selected immuno-
regulators of the immune response that contribute to regulatory genes, and that the IL-10 up-modulation in TS
the maintenance of intestinal immune homeostasis by mouse CD4+ T cells and the IL-10 down-modulation in
actively suppressing reactive responses to food antigens TR mouse CD4+ T cells are coherent mechanisms to
and commensal flora. Naive TS mice have higher CD25+ explain the naive and post-antigen feeding profiles of
and Foxp3+ mean frequencies among CD4+ lymphocytes both strains of mice.
than TR mice (Fig. 4). However, in TS and TR mice fed
with OVA the CD25+ frequencies were similar. Instead,
Acknowledgements
the significantly higher IL-10 level produced by T cells
in both naive and OVA feed TS mice is suggestive of We thank Dr Claudia G. F. Matta (North Fluminense
Treg cell participation in the discrimination of TS and State University) for kindly supplying IgY, Dr Gerlinde A.
TR immune responses. Treg cells are indispensable for P. B. Teixeira (Federal Fluminense University) for the nut
homeostasis and regulation of the immune system, and extracts, and Drs Cid Couto Chaves and Marcia Giesta
there is also a suggestion that multiple signals could (Rio de Janeiro State University) for care of the animal
confer resistance to Treg cell suppression.40 TR mice colonies. The research was supported by CAPES, CNPq,
produce higher prostaglandin E2, intercellular adhesion FAPERJ, FENORTE and FUJB (Brazil).

10  2010 Blackwell Publishing Ltd, Immunology


Naive mice and OVA oral tolerance
20 Mowat AM. Depletion of suppressor T cells by 20 deoxyguanosine abrogates tolerance
Disclosures in mice fed ovalbumin and permits the induction of intestinal delayed type hypersensi-
tivity. Immunology 1986; 58:179–84.
There is no conflict of interest. 21 Avrameas S. Coupling of enzymes to proteins with glutaraldehyde. Use of the conju-
gates for the detection of antigens and antibodies. Immunochemistry 1969; 6:43–52.
22 da Silva MF, da Costa SC, Ribeiro RC, Sant’Anna OA, da Silva AC. Independent
References genetic control of B- and T-cell tolerance in strains of mouse selected for extreme phe-
notypes of oral tolerance. Scand J Immunol 2001; 53:148–54.
1 Garside P, Mowat AM, Khoruts A. Oral tolerance in disease. Gut 1999; 44:137–42. 23 Rizzo LV, Morawetz RA, Miller-Rivero NE et al. IL-4 and IL-10 are both required for
2 Mowat AM. Oral tolerance and regulation of immunity to dietary antigens. In: Ogra the induction of oral tolerance. J Immunol 1999; 162:2613–22.
PL, ed. Handbook of Mucosal Immunology. London: Academic Press Inc, 1994:185–210. 24 Yanaba K, Bouaziz JD, Matsushita T, Tsubata T, Tedder TF. The development and
3 Friedman A, Weiner HL. Induction of anergy or active suppression following oral tol- function of regulatory B cells expressing IL-10 (B10 cells) requires antigen receptor
erance is determined by antigen dosage. Proc Natl Acad Sci U S A 1994; 91:6688–92. diversity and TLR signals. J Immunol 2009; 182:7459–72.
4 Faria AM, Weiner HL. Oral tolerance and TGF-beta-producing cells. Inflamm Allergy 25 Biozzi G, Ribeiro OG, Saran A et al. Effect of genetic modification of acute inflamma-
Drug Targets 2006; 5:179–90. tory responsiveness on tumorigenesis in the mouse. Carcinogenesis 1998; 19:337–46.
5 Piccirillo CA, Letterio JJ, Thornton AM, McHugh RS, Mamura M, Mizuhara H, Shev- 26 Tobagus IT, Thomas WR, Holt PG. Adjuvant costimulation during secondary antigen
ach EM. CD4+ CD25+ regulatory T cells can mediate suppressor function in the challenge directs qualitative aspects of oral tolerance induction, particularly during the
absence of transforming growth factor beta1 production and responsiveness. J Exp Med neonatal period. J Immunol 2004; 172:2274–85.
2002; 196:237–46. 27 Conde AA, Stransky B, Faria AM, Vaz NM. Interruption of recently induced immune
6 Miller A, Lider O, Weiner H. Antigen-driven bystander suppression after oral adminis- responses by oral administration of antigen. Braz J Med Biol Res 1998; 31:377–80.
tration of antigens. J Exp Med 1991; 74:791–8. 28 Crispe IN. Hepatic T cells and liver tolerance. Nat Rev Immunol 2003; 3:51–62.
7 Faria AM, Weiner HL. Oral tolerance: mechanisms and therapeutic applications. Adv 29 Silva MFS, Nóbrega A, Ribeiro RC, Levy MS, Ribeiro OG, Tambourgi DV, Sant’Anna
Immunol 1999; 73:153–264. OA, Silva AC. Genetic selection for resistance or susceptibility to oral tolerance imparts
8 Mayer L, Shao L. Therapeutic potential of oral tolerance. Nat Rev Immunol 2004; correlation to both IgE level and mast cell number phenotypes with a profound impact
4:407–19. on the atopic potential of the individual. Clin Exp Allergy 2006; 36:1399–407.
9 Maeda Y, Noda S, Tanaka K et al. The failure of oral tolerance induction is function- 30 Tavares DAG, Ribeiro RC, Silva AC. Inflammatory lesion and parasite load are inver-
ally coupled to the absence of T cells in Peyer’s patches under germ-free conditions. sely associated in Leishmania amazonensis infected mice genetically selected according
Immunobiology 2001; 204:442–57. to oral tolerance susceptibility. Microbes Infect 2006; 8:957–64.
10 Bilsborough J, Viney JL. Getting to the guts of immune regulation. Immunology 2002; 31 Slavin AJ, Maron R, Weiner HL. Mucosal administration of IL-10 enhances oral toler-
106:139–43. ance in autoimmune encephalomyelitis and diabetes. Int Immunol 2001; 13:825–33.
11 Kraus TA, Cheifetz A, Toy L, Meddings JB, Mayer L. Evidence for a genetic defect in 32 Dieckmann D, Bruett CH, Ploettner H, Lutz MB, Schuler G. Human CD4+ CD25+ reg-
oral tolerance induction in inflammatory bowel disease. Inflamm Bowel Dis 2006; ulatory, contact-dependent T cells induce interleukin 10-producing, contact-indepen-
12:82–8. dent type 1-like regulatory T cells. J Exp Med 2002; 196:247–53.
12 Vaz NM, Rios MJ, Lopes LM, Gontijo CM, Castanheira EB, Jacquemart F, Andrade LA. 33 Inobe J, Slavin AJ, Komagata Y, Chen Y, Liu L, Weiner HL. IL-4 is a differentiation
Genetics of susceptibility to oral tolerance to ovalbumin. Braz J Med Biol Res 1987; factor for transforming growth factor-beta secreting Th3 cells and oral administration
20:785–90. of IL-4 enhances oral tolerance in experimental allergic encephalomyelitis. Eur J Immu-
13 Lamont AG, Mowat AM, Browning MJ, Parrott DM. Genetic control of oral tolerance nol 1998; 28:2780–90.
to ovalbumin in mice. Immunology 1988; 63:737–9. 34 Cottrez F, Groux H. Specialization in tolerance: innate CD4+ CD25+ versus acquired
14 da Silva AC, de Souza KW, Machado RC, da Silva MF, Sant’Anna OA. Genetics of Tr1 and Th3 regulatory T cells. Transplantation 2004; 77:S12–5.
immunological tolerance: I. Bidirectional selective breeding of mice for oral tolerance. 35 Glimcher LH, Murphy KM. Lineage commitment in the immune system: the T helper
Res Immunol 1998; 149:151–61. lymphocyte grows up. Genes Dev 2000; 14:1693–711.
15 McLaren RD, Prosser CG, Grieve RC, Borissenko M. The use of caprylic acid for the 36 Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, Weaver
extraction of the immunoglobulin fraction from egg yolk of chickens immunised with CT. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from
ovine alpha-lactalbumin. J Immunol Methods 1994; 177:175–84. the T helper type 1 and 2 lineages. Nat Immunol 2005; 6:1123–32.
16 Landry J, Moureaux T. Heterogeneity of corn seed glutelin: selective extraction and 37 Ouyang W, Kolls JK, Zheng Y. The biological functions of T helper 17 cell effector
amino acid composition of the 3 isolated fractions. Bull Soc Chim Biol (Paris) 1970; cytokines in inflammation. Immunity 2008; 28:454–67.
52:1021–37. 38 Mars LT, Araujo L, Kerschen P et al. Invariant NKT cells inhibit development of the
17 Lowry QH, Rodebrouch NJ, Farr AL, Randall J. Protein measurement with folin phenol Th17 lineage. Proc Natl Acad Sci U S A 2009; 106:6238–43.
reagent. Biol Chem (Bethesda) 1951; 193:265–75. 39 da Silva MF, da Nóbrega A, Barja-Fidalgo TC, Brando-Lima AC, Cunha F, da Silva AC.
18 Bruce MG, Ferguson A. Oral tolerance to ovalbumin in mice: studies of chemically Genetic selection for susceptibility to oral tolerance leads to a profound reduction of
modified and ‘biologically filtered’ antigen. Immunology 1986; 57:627–30. the acute inflammatory response. Ann N Y Acad Sci 2004; 1029:398–401.
19 Faria AM, Maron R, Ficker SM, Slavin AJ, Spahn T, Weiner HL. Oral tolerance 40 Walker LS. Regulatory T cells overturned: the effectors fight back. Immunology 2009;
induced by continuous feeding: enhanced up-regulation of transforming growth factor- 126:466–74.
beta/interleukin-10 and suppression of experimental autoimmune encephalomyelitis. J
Autoimmun 2003; 20:135–45.

 2010 Blackwell Publishing Ltd, Immunology 11

Vous aimerez peut-être aussi