ArticlePDF Available

A Novel Approach to Specific Allergy Treatment: The Recombinant Allergen-S-Layer Fusion Protein rSbsC-Bet v 1 Matures Dendritic Cells That Prime Th0/Th1 and IL-10-Producing Regulatory T Cells

Authors:

Abstract and Figures

An ideal vaccine for allergen-specific immunotherapy of type I allergies should display reduced mediator-releasing capacity, induce maturation of APC, and modify the disease-eliciting Th2-dominated allergen-specific response to a more physiological response. We have previously shown that rSbsC-Bet v 1, the recombinant fusion protein of a bacterial surface (S-layer) protein of Geobacillus stearothermophilus ATCC 12980 and the major birch pollen allergen Bet v 1, exhibited reduced allergenicity and induced IFN-gamma and IL-10 synthesis in Bet v 1-specific Th2 clones. In this study, we characterized the effects of rSbsC-Bet v 1 on immature monocyte-derived dendritic cells (mdDC) and the consequences for the polarization of naive CD4(+) T lymphocytes isolated from the blood of birch pollen-allergic patients. mdDC responded to rSbsC-Bet v 1 with a significant up-regulation of costimulatory molecules, functional maturation, and the synthesis of IL-10 and IL-12. mdDC matured with rSbsC-Bet v 1 induced the differentiation of naive T cells into IFN-gamma-producing cells. This effect was IL-12 dependent. In parallel, a substantial number of naive T cells developed into IL-10-producing CD25(+)Foxp3(+)CLTA-4(+) cells capable of active suppression. Thus, rSbsC-Bet v 1 showed immune stimulatory capacity on DC, which then promoted the simultaneous differentiation of Th0/Th1 cells and regulatory T cells. These data further support that the concept of conjugating allergens to bacterial agents is a promising approach to improve vaccines for specific immunotherapy of atopic allergies.
Content may be subject to copyright.
A Novel Approach to Specific Allergy Treatment: The
Recombinant Allergen-S-Layer Fusion Protein rSbsC-Bet v 1
Matures Dendritic Cells That Prime Th0/Th1 and
IL-10-Producing Regulatory T Cells
1
Marianne Gerstmayr,* Nicola Ilk,
Irma Schabussova,* Beatrice Jahn-Schmid,*
Eva M. Egelseer,
Uwe B. Sleytr,
Christof Ebner,
and Barbara Bohle
2
*
An ideal vaccine for allergen-specific immunotherapy of type I allergies should display reduced mediator-releasing capacity,
induce maturation of APC, and modify the disease-eliciting Th2-dominated allergen-specific response to a more physiological
response. We have previously shown that rSbsC-Bet v 1, the recombinant fusion protein of a bacterial surface (S-layer) protein
of Geobacillus stearothermophilus ATCC 12980 and the major birch pollen allergen Bet v 1, exhibited reduced allergenicity and
induced IFN-
and IL-10 synthesis in Bet v 1-specific Th2 clones. In this study, we characterized the effects of rSbsC-Betv1on
immature monocyte-derived dendritic cells (mdDC) and the consequences for the polarization of naive CD4
T lymphocytes
isolated from the blood of birch pollen-allergic patients. mdDC responded to rSbsC-Bet v 1 with a significant up-regulation of
costimulatory molecules, functional maturation, and the synthesis of IL-10 and IL-12. mdDC matured with rSbsC-Bet v 1 induced
the differentiation of naive T cells into IFN-
-producing cells. This effect was IL-12 dependent. In parallel, a substantial number
of naive T cells developed into IL-10-producing CD25
Foxp3
CLTA-4
cells capable of active suppression. Thus, rSbsC-Bet v
1 showed immune stimulatory capacity on DC, which then promoted the simultaneous differentiation of Th0/Th1 cells and
regulatory T cells. These data further support that the concept of conjugating allergens to bacterial agents is a promising approach
to improve vaccines for specific immunotherapy of atopic allergies. The Journal of Immunology, 2007, 179: 7270 –7275.
Allergen-specific Th2 cells, i.e., CD4
T lymphocytes
that produce high levels of IL-4, IL-5, and IL-13, but
low amounts of IFN-
, are pivotal for the induction and
maintenance of type I allergies (1). The only approved treatment
that counterregulates the disease-eliciting allergen-specific Th2 re-
sponse in allergic individuals is specific immunotherapy (SIT).
3
Successful SIT significantly reduces allergen-induced T cell pro-
liferation, indicating the induction of peripheral tolerance in aller-
gen-specific T cells. In parallel, the shift from Th2 toward a more
physiological Th0/Th1-like immune response, either by reduced
IL-4 production or the induction of IFN-
, has been observed
(2–5). More recently, the induction of IL-10-producing allergen-
specific T cells has been associated with successful SIT (6 –9).
SIT-induced IL-10-producing CD4
CD25
T cells suppress pro-
liferation and cytokine production of allergen-specific T cells and
are thus designated T regulatory (Treg) cells (10 –13). Further-
more, the fine balance between IL-10-producing allergen-specific
Treg and Th2 cells has been considered a relevant component of
the physiological immune response to allergens (14).
One strategy to improve vaccines for SIT is the use of adjuvants
that amplify the modulation of the allergen-specific Th2 response.
In this respect, different agents from bacterial origin have been
regarded as promising candidates. Synthetic lipopeptides derived
from lipoproteins of Gram-negative bacteria have been reported to
induce IFN-
and IL-10 synthesis by human T cells (15). Mono-
phosphoryl lipid A, a nontoxic derivate of LPS of Salmonella min-
nesota, mixed with grass pollen extract enhanced the production of
IFN-
and reduced the production of IL-5 in PBMC from grass
pollen-allergic patients (16). Synthetic oligodeoxynucleotides con-
taining CpG motifs (CpG-ODN) derived from bacterial DNA pro-
moted Th1-like cytokine production and decreased the spontane-
ous synthesis of IgE Abs in allergic individuals in vitro (17–20).
This immunomodulatory capacity was even more pronounced
when CpG-ODN were chemically linked to a single defined aller-
gen (21–23). Both conjugated CpG-ODN and monophosphoryl
lipid A have been successfully applied in clinical trials and re-
vealed promising results regarding the treatment of type I allergies
(24 –26).
S-layer proteins forming crystalline arrays on the cell surface of
many bacteria and most archaea have already a long tradition in
vaccine development (27–29). We have proposed to engineer al-
lergy vaccines by genetic fusion of allergens with S-layer proteins:
*Department of Pathophysiology, Center for Physiology, Pathophysiology, and Im-
munology, Medical University of Vienna, Vienna, Austria; and
Center for Nano-
Biotechnology, University of Natural Resources and Applied Life Sciences and
Al-
lergy Clinic Reumannplatz, Vienna, Austria
Received for publication July 10, 2007. Accepted for publication September 19, 2007.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by the Fonds zur Fo¨rderung der wissenschaftlichen For-
schung (SFB-F1807-B04); P12158 by the OeNB, Austria; P18510-B12 by the Euro-
pean Union Project NAS-SAP; and by the U.S. Air Force Office of Scientific Re-
search, Project FA9550-07-1-0313.
2
Address correspondence and reprint requests to Dr. Barbara Bohle, Department of
Pathophysiology, Center for Physiology, Pathophysiology, and Immunology, Medical
University of Vienna, Austria; AKH-3Q, Waehringer Guertel 18-20, Vienna, Austria.
E-mail address: barbara.bohle@meduniwien.ac.at
3
Abbreviations used in this paper: SIT, specific immunotherapy; DC, dendritic cell;
mdDC, monocyte-derived DC; B-DC, mdDC incubated with rBet v 1; CpG-ODN,
oligodeoxynucleotides containing CpG motifs; Ct, threshold cycle; LB-CD, mdDC
incubated with LPS and rBet v 1; SB-DC, mdDC incubated with rSbsC-Bet b 1; Treg,
T regulatory.
Copyright © 2007 by The American Association of Immunologists, Inc. 0022-1767/07/$2.00
The Journal of Immunology
www.jimmunol.org
the major birch pollen allergen Bet v 1 was fused with the S-layer
protein from the nonpathogenic bacteria Geobacillus stearother-
mophilus ATCC 12980 (30, 31). The resulting allergen-S-layer
fusion protein rSbsC-Bet v 1 induced IFN-
and IL-10 production
in PBMC and Bet v 1-specific Th2 clones from birch pollen-al-
lergic donors (31). Moreover, rSbsC-Bet v 1 displayed a reduced
IgE-binding capacity, and consequently a lower risk to induce ad-
verse vaccine-related anaphylactic reactions (31). Thus, we con-
sider rSbsC-Betv1aspromising hypoallergenic molecule to skew
allergen-specific Th2 effector cells to both IFN-
and IL-10 pro-
duction. In this study, we investigated whether rSbsC-Betv1af-
fected the differentiation of naive CD4
T cells from birch pollen-
allergic individuals. We first analyzed the effects of rSbsC-Bet v 1
on monocyte-derived dendritic cells (mdDC) regarding matura-
tion, functionality, and cytokine production. Next, mdDC stimu-
lated with rSbsC-Bet v 1 were used to prime naive CD4
T cells.
T cell polarization was evaluated by measuring IL-4, IFN-
, and
IL-10. The suppressive capacity of IL-10-secreting T cells was
tested in proliferation assays.
Materials and Methods
Patients, allergens, and reagents
Birch pollen-allergic patients with a typical case history, positive skin prick
tests, and CAP/RAST scores 3 (Pharmacia) to birch were included. Sen-
sitization to Bet v 1 was assessed by means of immunoblotting. The study
was approved by the local medical ethics committee (Vienna, Austria).
RSbsC-Bet v 1 and rSbsC were produced, as described (30). Endotoxin
levels in rSbsC-Bet v 1 and rSbsC were below 0.4 EU/mg, as determined
by Limulus amebocyte lysate assay (BioWhittaker). RBet v 1 was pur-
chased from Biomay, and LPS from Sigma-Aldrich.
Generation of mdDC
Monocytes were isolated from PBMC by magnetic cell sorting using anti-
CD14 microbeads (Miltenyi Biotec). The isolated fraction contained
95% CD14
cells, as determined by flow cytometry using FITC-conju-
gated anti-CD14 mAb (BD Pharmingen). CD14
cells (1 10
6
/ml) were
cultured in 24-well plates (Corning Glass) in RPMI 1640 medium (Invitro-
gen Life Technologies) supplemented with 2 mM L-glutamine, 170 mg/L
gentamycinsulfate (both Sigma-Aldrich), human rIL-4 (1000 U/ml; Strath-
mann Biotec), and GM-CSF (50 ng/ml; PeproTech) in the presence of
either 3% autologous plasma or 10% heat-inactivated FCS (PAA Labora-
tories) for 7 days. The resulting dendritic cell (DC) population was homo-
geneously CD14
, and 70% of the cells reacted with PE-Cy5-conjugated
anti-CD1a mAbs (BD Pharmingen). Immature DC were either left un-
treated or stimulated with rBetv1(10
g/ml), rSbsC-Betv1(59
g/ml
containing 10
g/ml rBet v 1), and corresponding amounts of rSbsC and
LPS (1
g/ml), respectively. After 48 h, surface expression of DC matu-
ration markers on CD14
CD1a
cells was analyzed using PE-conjugated
anti-CD80, anti-CD83 mAb (both from BD Pharmingen), anti-CD86 mAb
(Caltag Laboratories), and FITC-conjugated anti-CD40 mAb (provided by
O. Majdic, Institute of Immunology, Vienna, Austria).
Mixed leukocyte reaction
Differently stimulated DC were harvested after 48 h, irradiated (60 Gy),
and added to 1 10
5
allogeneic PBMC from nonallergic donors in dif-
ferent ratios. Cells were cultured in triplicates in 96-well plates (Nunclone;
Nunc) in serum-free UC medium (BioWhittaker) supplemented with 2 mM
L-glutamine and 2 10
5
M 2-ME for 6 days. Proliferation was assessed
by adding [
3
H]thymidine (0.5
Ci/well) during the last 16 h of culture and
measuring the incorporated radioactivity by scintillation counting.
Real-time PCR
DC were incubated in 24-well plates (Corning Glass) with different stimuli
for 14 h. RNA was isolated using the RNeasy Mini Kit (Qiagen) and
reversely transcribed with TaqMan reverse-transcription reagents using
random hexamers (Applied Biosystems). Analysis of 18S rRNA (control
housekeeping gene), IL-10, and IL-12p40 was performed with cDNA-spe-
cific Assays-on-Demand (Applied Biosystems) using an ABI PRISM 7700
Sequence Detection System (Applied Biosystems). A negative control with-
out template and a positive control with a template of known amplification
were included in each run. All amplifications were performed in triplicates.
Relative quantification and calculation of the range of confidence were per-
formed by using the comparative threshold cycle (⌬⌬Ct) method (Applied
Biosystems). The fold induction of cytokines was calculated by the formula
2
⫺⌬⌬CT
2
(Ct for unstimulated culture ⫺⌬Ct for stimulated culture)
.
Purification and stimulation of naive CD4
T cells
Peripheral CD4
T cells were purified using the CD4 Negative Isolation
Kit from Dynal Biotech. From the resulting CD4
population, CD45RA
cells were isolated by magnetic cell sorting using anti-CD45RA mi-
crobeads (Miltenyi Biotec). A purity of 92% CD4
CD45RA
T cells
**
*
Mean fluorescence intensity
200
150
100
50
0
CD40
Percent positive cells
100
80
60
40
20
0
CD80
100
80
60
40
20
0
CD83
100
80
60
40
20
0
CD86
**
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
**
**
**
*
**
*
*
**
*
*
*
**
**
***
**
Mean fluorescence intensity
200
150
100
50
0
CD40
Percent positive cells
100
80
60
40
20
0
CD80
100
80
60
40
20
0
CD83
100
80
60
40
20
0
CD86
** **
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
Medium
rBet v1
rSbsC-Bet v1
rSbsC
LPS
****
*
***
** *
*
**
*
***
**
**
**
**
***
**
*
*
**
FIGURE 1. rSbsC-Bet v 1 induces
DC maturation. Immature mdDC were
left untreated or stimulated with rBet v
1, rSbsC-Bet v 1, rSbsC, or LPS. The
percentage of DC expressing CD80,
CD83, and CD86 surface markers and
the mean fluorescence intensity of
CD40 are shown. Data from seven
different birch pollen-allergic donors
were summarized in box plots. Each
box represents the interquartile range
containing 50% of the data. The line
across the box indicates the median.
The significances of differences to rBet
v 1 were calculated using the Wilcoxon
signed ranks test (,p0.05).
0
30
60
90
120
150
180
2510
x10³ DC/well
Proliferation (cpm x103)
Medium rBet v 1
rSbsC-Bet v1 rSbsC
LPS
0
30
60
90
120
150
180
2510
x10³ DC/well
Proliferation (cpm x103)
Medium rBet v 1
rSbsC-Bet v1 rSbsC
LPS
FIGURE 2. rSbsC-Bet v 1 induces functional DC. Increasing numbers
of untreated mdDC or mdDC incubated with rBet v 1, rSbsC-Bet v 1,
rSbsC, or LPS were cultured with 1 10
5
allogeneic PBMC. Mean cpm
of triplicates of one experiment of three are shown.
7271The Journal of Immunology
was achieved. Autologous immature DC were stimulated with either rBet
v 1 (10
g/ml), rSbsC-Betv1(59
g/ml), or LPS (100 ng/ml) plus rBet
v 1 (10
g/ml). After 24 h, 1 10
6
CD4
CD45RA
T cells were added
(day 0). At day 7, T cells were restimulated with the respective DC pop-
ulation and analyzed for cytokine production and proliferation at day 14. In
some experiments, a neutralizing anti-IL-12 mAb (50
g/ml; R&D Sys-
tems) was added at days 0 and 7.
Phenotyping of primed CD4
T cells
T cells were stained with PE-conjugated anti-CD25, PE-Cy5-conjugated
anti-CD152 mAb (both from BD Pharmingen), and a FITC-conjugated
anti-human Foxp3 (eBioscience). Intracellular cytokine detection in T cells
was performed using FITC-, PE-, PerCP-labeled anti-IFN-
, anti-IL-4, anti-
CD3 mAbs, and respective isotype control Abs, as described (31). To quan-
tify cytokine levels, 1 10
5
T cells were stimulated in triplicates with
anti-CD3 (1
g/ml OKT3) and anti-CD28 (1
g/ml; Sanquin) in the pres-
ence of irradiated PBMC (60 Gy) in 96-well plates for 48 h. Levels of
IL-10, IFN-
, and TGF-
in supernatants were analyzed by ELISA using
matched Ab pairs (Pierce) and a TGF-
1 ELISA kit (IBT) (31).
Evaluation of suppressive capacity of IL-10-secreting T cells
Purified CD4
T cells (1 10
6
) were stimulated with immobilized anti-
CD3 (0.5
g/ml) and soluble anti-CD28 (1
g/ml) in 48-well plates. After
18 h, IL-10-secreting cells were isolated using the IL-10 Secretion Assay-
Cell Enrichment and Detection Kit (Milteny Biotec), according to the man-
ufacturers protocol. Autologous CD4
CD25
cells were cocultured with
IL-10-secreting cells in different ratios in the presence of anti-CD3 (OKT3
0.5
g/ml) or without any stimulus. After 72 h, proliferation was assessed.
Statistical analysis
Statistical significance of differences was determined using the Wilcoxon
signed ranks test. Differences were considered statistically significant for
p0.05.
Results
rSbsC-Bet v 1 induces phenotypic and functional maturation
of DC
Immature mdDC from seven different birch pollen-allergic patients
were left untreated or incubated with rBet v 1, rSbsC-Bet v 1, and
rSbsC for 48 h (Fig. 1). As positive control, LPS, a well-charac-
terized agent to mature mdDC, was used. Incubation of mdDC
with rBet v 1 showed no effects as compared with medium alone.
Incubation with rSbsC-Bet v 1 and rSbsC induced a significant
up-regulation of CD80 ( p0.018, p0.018), CD83 (p0.018,
p0.018), CD86 ( p0.018, p0.018), and CD40 ( p0.018,
p0.028) as compared with rBet v 1 (Fig. 1). RSbsC-Bet v 1
induced a significantly higher expression of CD83 than rSbsC
(p0.043) (Fig. 1). LPS-stimulated mdDC displayed signifi-
cantly higher levels of all surface markers as compared with
rSbsC-Bet v 1 (CD80, p0.018; CD83, p0.043; CD86, p
0.018; CD40, p0.018). To test whether the differently matured
mdDC were capable of activating T cells, MLR were performed.
The allostimulatory capacity of mdDC reflected the observed
changes of surface marker expression (Fig. 2). In comparison with
unstimulated or rBet v 1-stimulated mdDC, mdDC incubated with
rSbsC-Betv1orrSbsC induced enhanced proliferative responses
in allogeneic T cells. LPS-stimulated mdDC induced a more pro-
nounced T cell proliferation as compared with rSbsC-Bet v 1
(Fig. 2).
rSbsC-Bet v 1 induces IL-10 and IL-12 production in DC
IL-10 and IL-12 mRNA expression levels in response to the dif-
ferent stimuli were assessed by real-time PCR in mdDC generated
from six different birch pollen-allergic donors (Fig. 3). The me-
dium control was used as baseline for calculating the fold induc-
tion of IL-10 and IL-12, respectively. The major birch pollen al-
lergen marginally enhanced IL-10 and IL-12 mRNA expression in
mdDC. Compared with rBet v 1, rSbsC-Bet v 1 induced a signif-
icantly higher mRNA expression for both cytokines ( p0.028,
p0.028) as did LPS ( p0.046, p0.028; Fig. 3), and stim-
ulation with rSbsC resulted in a significant increase of IL-12
mRNA expression ( p0.046). These results obtained at the
mRNA level were confirmed by ELISA of supernatants derived
from mdDC stimulated for 48 h (data not shown).
IL-10
rBet v1
rSbsC-Betv1
rSbsC
LPS
0
2
4
6
8
10
12
14
16 *
***
IL-10 mRNA (fold induction)
IL-12
rBet v1
rSbsC-Bet v 1
rSbsC
LPS
0
10
20
30
40
50
60 *
*
**
IL-12 mRNA (fold induction)
IL-10
rBet v1
rSbsC-Betv1
rSbsC
LPS
0
2
4
6
8
10
12
14
16 *
***
IL-10 mRNA (fold induction)
IL-12
rBet v1
rSbsC-Bet v 1
rSbsC
LPS
0
10
20
30
40
50
60 *
*
**
IL-12 mRNA (fold induction)
FIGURE 3. rSbsC-Bet v 1 induces IL-10 and IL-12
in DC. Immature mdDC from six patients were left un-
treated or stimulated with rBet v 1, rSbsC-Bet v 1,
rSbsC, or LPS. The relative expression of IL-10 and
IL-12 mRNA was individually calculated as compared
with the mRNA expression in unstimulated cells. Lines
indicate the median. ,p0.05 (Wilcoxon signed ranks
test).
FIGURE 4. DC matured with rSbsC-Bet v 1 promote a Th0/Th1-like
response. Naive CD4
T cells were primed with autologous B-DC, LB-
DC, and SB-DC in the absence or presence of anti-IL-12. Intracellular
synthesis of IFN-
and IL-4 was determined. Three experiments (patients
I–III) of 10 are shown.
7272 EFFECTS OF rSbsC-Betv1ONDCANDNAIVE CD4
T CELLS
rSbsC-Bet v 1-matured DC promote IL-12-dependent Th0/Th1
differentiation of naive CD4
T cells
CD4
CD45RA
cells derived from 10 birch pollen-allergic pa-
tients were stimulated twice with autologous mdDC incubated
with either rBet v 1 (B-DC), rSbsC-Bet v 1 (SB-DC), or simulta-
neous addition of LPS and rBet v 1 (LB-DC). T cell polarization
was determined by intracellular cytokine detection (Fig. 4). Com-
pared with B-DC, both SB-DC and LB-DC induced significantly
higher numbers of IFN-
-producing T cells ( p0.005; p
0.013) and enhanced numbers of IL-4-producing T cells. Similar to
LB-DC, SB-DC induced significantly more IFN-
than IL-4
T
cells ( p0.005), reflecting a Th1-like response. Interestingly,
significantly higher numbers of IL-4
IFN-
double-positive T
cells ( p0.017) were detected in SB-DC-primed T cells as com-
pared with LB-DC-stimulated cultures. The simultaneous addition
of neutralizing anti-IL-12 mAb and SB-DC to CD4
CD45RA
cells drastically reduced the number of IFN-
-producing CD4
T
cells (Fig. 4).
rSbsC-Bet v 1-matured DC promote the differentiation of
regulatory T cells
To investigate whether the different DC maturation induced by
rSbsC-Bet v 1 and LPS plus rBet v 1 translated into different Th
responses, the production of IFN-
, IL-10, and TGF-
was quan-
tified in supernatants of anti-CD3-stimulated T cells primed with
the differently stimulated mdDC by ELISA (Fig. 5). Compared
with B-DC-primed T cells, SB-DC- as well as LB-DC-primed T
cells produced significantly higher levels of IFN-
(p0.037;
p0.047) and IL-10 ( p0.005; p0.009). SB-DC-primed T
cells synthesized comparable levels of IFN-
and significantly
higher amounts of IL-10 ( p0.022) and TGF-
(p0.028) than
LB-DC-primed T cells. Because these cytokines are typically syn-
thesized by Treg cells, T cells from six individuals were stained for
CD25 and intracellular expression of Foxp3 7 days after the sec-
ond addition of DC. SB-DC-primed T cell cultures contained sig-
nificantly higher numbers of CD25
Foxp3
cells than LB-DC-
primed T cells ( p0.028). One representative experiment is
shown in Fig. 6A. Parallel staining for CTLA-4 revealed that
CD25
Foxp3
cells were all CTLA-4 positive (data not shown).
To analyze whether IL-10-producing cells expressed these Treg
markers, T cells needed to be stimulated before intracellular cy-
tokine detection. Most T cells became Foxp3 positive in response
to PMA/ionomycin, whereas the expression of CTLA-4 was hardly
affected (data not shown). Therefore, we costained T cells for
IL-10 and CTLA-4 and observed that all IL-10
cells were CTLA-
4
. One representative example is shown in Fig. 6B. Next, T cells
from four individuals were stained for their anti-CD3-induced se-
cretion of IL-10 and IFN-
. The majority of IL-10-secreting cells
constituted a population different from IFN-
-secreting cells, but
also cells secreting both cytokines were detected. One example is
depicted in Fig. 6C. Subsequently, we isolated IL-10-secreting
cells from SB-DC-primed T cell cultures from two patients. Pro-
liferative responses of these cells were markedly lower as com-
pared with autologous CD4
CD25
cells (Fig. 6D). Coculturing
IL-10-secreting T cells with autologous CD4
CD25
cells in the
presence of irradiated autologous PBMC and anti-CD3 Abs re-
sulted in the suppression of the proliferation of CD4
CD25
cells
in a dose-dependent manner (Fig. 6D). In two independent exper-
iments, a mean suppression of 83% was observed at a suppressor:
responder cell ratio of 1:1 and 74% at a ratio of 1:3, respectively.
Discussion
We have previously shown that genetic fusion of the major birch
pollen allergen and a bacterial S layer resulted in a recombinant
protein exhibiting reduced mediator-releasing capacity, retained
pg/ml
3000
2500
2000
1500
1000
500
0
*
*
IFN-gamma IL-10 TGF-beta
600
500
400
300
200
100
0
*
*
*
160
140
120
100
80
60
40
20
0
B-DC
LB-DC
SB-DC
B-DC
LB-DC
SB-DC
B-DC
LB-DC
SB-DC
*
pg/ml
3000
2500
2000
1500
1000
500
0
**
**
IFN-gamma IL-10 TGF-beta
600
500
400
300
200
100
0
**
**
**
160
140
120
100
80
60
40
20
0
B-DC
LB-DC
SB-DC
B-DC
LB-DC
SB-DC
B-DC
LB-DC
SB-DC
B-DC
LB-DC
SB-DC
B-DC
LB-DC
SB-DC
B-DC
LB-DC
SB-DC
**
FIGURE 5. T cells primed with mdDC matured with rSbsC-Bet v 1
produce IFN-
, IL-10, and TGF-
. Naive CD4
T cells were primed with
autologous B-DC, LB-DC, and SB-DC. After stimulation with anti-CD3
plus anti-CD28, supernatants were assessed for cytokines by ELISA. Data
from 10 different donors were summarized in box plots. ,p0.05 (Wil-
coxon signed ranks test).
FIGURE 6. rSbsC-Bet v 1-matured mdDC prime IL-
10-producing Treg cells. Naive CD4
T cells were
primed with autologous LB-DC or SB-DC and stained
for A, CD25 and intracellular Foxp3; B, intracellular
IL-10 and CTLA-4; and C, secretion of IL-10 and
IFN-
assessed by cytokine secretion assays. One rep-
resentative experiment of each staining is shown. D,
SB-DC-induced IL-10-secreting cells were isolated
from two donors and cocultured with autologous
CD4
CD25
cells in the presence of irradiated autol-
ogous PBMC and anti-CD3. Mean cpm and SD of trip-
licates are depicted.
7273The Journal of Immunology
Bet v 1-T cell epitopes, and the potency to induce IFN-
and IL-10
production in Bet v 1-specific Th2 clones (31). In this study, we
analyzed the effects of rSbsC-Betv1onimmature DC and naive
CD4
T cells isolated from the blood of birch pollen-allergic pa-
tients. We demonstrate that DC matured with rSbsC-Bet v 1 pro-
mote the simultaneous differentiation of Th0/Th1 cells and IL-10-
producing Treg cells. These data further support the concept that
recombinant fusion of allergens and S-layer proteins is a promising
approach to improve vaccines for SIT of atopic allergies.
In a first step, the effects of rSbsC-Bet v 1 and rSbsC on im-
mature mdDC were analyzed. Stimulation with these proteins re-
sulted in DC maturation, increased T cell stimulatory property, and
the production of IL-10 and IL-12 (Figs. 1–3). Thus, the recom-
binant S-layer protein from G. stearothermophilus ATCC 12980
stimulates the innate immune system of allergic patients. These
findings are in line with the effects of natural S layers isolated from
Clostridium difficile on monocytes and mdDC of nonallergic indi-
viduals (32). Stimulation of DC with rBet v 1 alone resulted nei-
ther in activation nor maturation, indicating that an additional stim-
ulus is required to induce strong immune responses to the
recombinant allergen. This activity was introduced by recombinant
fusion of rBetv1totheS-layer protein. The fact that rSbsC-Bet
v 1 displayed stronger effects than rSbsC may be referred to the
involvement of Ab receptor-mediated uptake of rSbsC-Bet v 1. We
have previously shown that the fusion protein contains relevant
IgE-binding epitopes of Bet v 1 (31). It may well be possible that
rSbsC-Bet v 1 was recognized by Bet v 1-specific IgE Abs bound
to the high affinity receptor FcRI on APC, resulting in IgE-me-
diated uptake in addition to phagocytosis (33).
In a second step, we used rSbsC-Bet v 1-matured DC (SB-DC)
to prime naive CD4
T cells from birch pollen-allergic donors and
investigated the polarization of the T cell response. To induce an
allergen-specific Th1 response, mdDC were primed with rBet v 1
plus LPS (LB-DC). Similar to this positive control, priming of T
cells with SB-DC resulted in significantly more IFN-
than
IL-4
cells (Fig. 4). In line with our previous data showing that
S-layer-induced IFN-
synthesis in effector T cells was mediated
by IL-12 (34), SB-DC-induced T cell polarization toward IFN-
production also depended on the presence of this cytokine (Fig. 4).
Interestingly, significantly higher numbers of IL-4
IFN-
dou-
ble-positive T cells were detected in SB-DC-primed T cells as
compared with LB-DC-primed T cells. These cells may reflect a
Th0 phenotype, simultaneously synthesizing Th1 and Th2 cyto-
kines. Thus, rSbsC-Bet v 1 may have the advantage of not pro-
moting exclusive Th1-like immune responses. Nevertheless, po-
tential risks of in vivo administration of rSbsC-Bet v 1 need to be
carefully evaluated.
SB-DC primed naive CD4
T cells to synthesize significantly
more IL-10 and TGF-
as compared with LB-DC (Fig. 5). Fur-
thermore, higher numbers of IL-10
CTLA-4
Foxp3
CD25
cells were detected in SB-DC-primed cultures (Fig. 6, A–C).
Coculturing IL-10-secreting T cells isolated from SB-DC-primed
T cells suppressed proliferative responses of autologous
CD4
CD25
T cells (Fig. 6D). Together, these data provide
strong evidence that SB-DC primed naive T cells toward a popu-
lation of IL-10-producing Treg cells in addition to Th0/Th1 cells.
Preliminary studies in our laboratory indicated that rSbsC-Bet v 1
targets TLR2, because HEK293 cells stably transfected with TLR2
produced IL-8 in response to rSbsC-Bet v 1, whereas HEK293
cells transfected with TLR1 or TLR3–9 did not (data not shown).
The involvement of TLR2 may promote Treg cells through the
induction of IL-10 production in DC (35, 36). However, to date,
we observed comparable IL-10 levels produced by DC stimulated
with rSbsC-Betv1orLPSatthemRNA (Fig. 3) and protein level.
In contrast, the significantly lower expression of CD40, CD80, and
CD86 on the surface of SB-DC as compared with LB-DC may
indicate that rSbsC-Bet v 1 does not induce a fully matured DC
phenotype (Fig. 1). Immature and semimature DC have been dem-
onstrated to induce IL-10-producing Treg cells (37–39). Whether
other potential influences, e.g., enhanced expression of inhibitory
molecules of the B7 family or DC-derived TGF-
, are involved in
rSbsC-Bet v 1-induced development of Treg cells is currently un-
der investigation.
Our previous (31) and present data strongly suggest that rSbsC-
Bet v 1 meets several desired requirements for an ideal allergy
vaccine: less IgE-binding and reduced mediator-releasing capac-
ity; immune stimulatory effects on DC; immune modulatory effects
on naive and allergen-specific effector T cells; and a constant ratio
between allergen and adjuvant that should improve consistency of
the product used for SIT. In addition to the induction of a Th0/
Th1-like T cell response, rSbsC-Bet v 1 also promoted IL-10-pro-
ducing Treg cells. Both immune deviation and increased numbers
of circulating IL-10-secreting CD4
CD25
cells have been asso-
ciated with successful allergen SIT. Our in vitro data indicate that
rSbsC-Bet v 1 will simultaneously support these two immune
mechanisms. Due to its hypoallergenic features (31), this fusion
molecule may also reduce the risk of IgE-mediated side effects.
Therefore, allergens genetically fused to S-layer proteins are prom-
ising future vaccines for SIT of atopic allergies.
Acknowledgments
We thank Astrid Radakovics for excellent technical assistance. This work
is dedicated to Margit Sa´ra.
Disclosures
The authors have no financial conflict of interest.
References
1. Romagnani, S. 2000. T-cell subsets (Th1 versus Th2). Ann. Allergy Asthma Im-
munol. 85: 9 –18.
2. Secrist, H., C. J. Chelen, Y. Wen, J. D. Marshall, and D. T. Umetsu. 1993.
Allergen immunotherapy decreases interleukin 4 production in CD4
T cells
from allergic individuals. J. Exp. Med. 178: 2123–2130.
3. Durham, S. R., S. Ying, V. A. Varney, M. R. Jacobson, R. M. Sudderick,
I. S. Mackay, A. B. Kay, and Q. A. Hamid. 1996. Grass pollen immunotherapy
inhibits allergen-induced infiltration of CD4
T lymphocytes and eosinophils in
the nasal mucosa and increases the number of cells expressing messenger RNA
for interferon-
.J. Allergy Clin. Immunol. 97: 1356 –1365.
4. Jutel, M., W. J. Pichler, D. Skrbic, A. Urwyler, C. Dahinden, and U. R. Muller.
1995. Bee venom immunotherapy results in decrease of IL-4 and IL-5 and in-
crease of IFN-
secretion in specific allergen-stimulated T cell cultures. J. Im-
munol. 154: 4187– 4194.
5. Ebner, C., U. Siemann, B. Bohle, M. Willheim, U. Wiedermann, S. Schenk,
F. Klotz, H. Ebner, D. Kraft, and O. Scheiner. 1997. Immunological changes
during specific immunotherapy of grass pollen allergy: reduced lymphoprolifera-
tive responses to allergen and shift from TH2 to TH1 in T-cell clones specific for
Phl p 1, a major grass pollen allergen. Clin. Exp. Allergy 27: 1007–1015.
6. Bellinghausen, I., G. Metz, A. H. Enk, S. Christmann, J. Knop, and J. Saloga.
1997. Insect venom immunotherapy induces interleukin-10 production and a
Th2- to-Th1 shift, and changes surface marker expression in venom-allergic sub-
jects. Eur. J. Immunol. 27: 1131–1139.
7. Akdis, C. A., T. Blesken, M. Akdis, B. Wuthrich, and K. Blaser. 1998. Role of
interleukin 10 in specific immunotherapy. J. Clin. Invest. 102: 98 –106.
8. Francis, J. N., S. J. Till, and S. R. Durham. 2003. Induction of IL-
10
CD4
CD25
T cells by grass pollen immunotherapy. J. Allergy Clin. Im-
munol. 111: 1255–1261.
9. Nouri-Aria, K. T., P. A. Wachholz, J. N. Francis, M. R. Jacobson, S. M. Walker,
L. K. Wilcock, S. Q. Staple, R. C. Aalberse, S. J. Till, and S. R. Durham. 2004.
Grass pollen immunotherapy induces mucosal and peripheral IL-10 responses
and blocking IgG activity. J. Immunol. 172: 3252–3259.
10. Jutel, M., M. Akdis, F. Budak, C. Aebischer-Casaulta, M. Wrzyszcz, K. Blaser,
and C. A. Akdis. 2003. IL-10 and TGF-
cooperate in the regulatory T cell
response to mucosal allergens in normal immunity and specific immunotherapy.
Eur. J. Immunol. 33: 1205–1214.
11. Gardner, L. M., F. C. Thien, J. A. Douglass, J. M. Rolland, and R. E. O’Hehir.
2004. Induction of T ‘regulatory’ cells by standardized house dust mite immu-
notherapy: an increase in CD4
CD25
interleukin-10
T cells expressing pe-
ripheral tissue trafficking markers. Clin. Exp. Allergy 34: 1209 –1219.
7274 EFFECTS OF rSbsC-Betv1ONDCANDNAIVE CD4
T CELLS
12. Reefer, A. J., R. M. Carneiro, N. J. Custis, T. A. Platts-Mills, S. S. Sung,
J. Hammer, and J. A. Woodfolk. 2004. A role for IL-10-mediated HLA-DR7-
restricted T cell-dependent events in development of the modified Th2 response
to cat allergen. J. Immunol. 172: 2763–2772.
13. Bohle, B., T. Kinaciyan, M. Gerstmayr, A. Radakovics, B. Jahn-Schmid, and
C. Ebner. 2007. Sublingual immunotherapy induces IL-10-producing T regula-
tory cells, allergen-specific T-cell tolerance, and immune deviation.
J. Allergy Clin. Immunol. 120: 707–713.
14. Akdis, M., J. Verhagen, A. Taylor, F. Karamloo, C. Karagiannidis, R. Crameri,
S. Thunberg, G. Deniz, R. Valenta, H. Fiebig, et al. 2004. Immune responses in
healthy and allergic individuals are characterized by a fine balance between al-
lergen-specific T regulatory 1 and T helper 2 cells. J. Exp. Med. 199: 1567–1575.
15. Akdis, C. A., F. Kussebi, B. Pulendran, M. Akdis, R. P. Lauener, C. B.
Schmidt-Weber, S. Klunker, G. Isitmangil, N. Hansjee, T. A. Wynn, et al. 2003.
Inhibition of T helper 2-type responses, IgE production and eosinophilia by syn-
thetic lipopeptides. Eur. J. Immunol. 33: 2717–2726.
16. Puggioni, F., S. R. Durham, and J. N. Francis. 2005. Monophosphoryl lipid A
(MPL) promotes allergen-induced immune deviation in favor of Th1 responses.
Allergy 60: 678 684.
17. Parronchi, P., F. Brugnolo, F. Annunziato, C. Manuelli, S. Sampognaro,
C. Mavilia, S. Romagnani, and E. Maggi. 1999. Phosphorothioate oligode-
oxynucleotides promote the in vitro development of human allergen-specific
CD4
T cells into Th1 effectors. J. Immunol. 163: 5946 –5953.
18. Bohle, B., B. Jahn-Schmid, D. Maurer, D. Kraft, and C. Ebner. 1999. Oligode-
oxynucleotides containing CpG motifs induce IL-12, IL-18 and IFN-
production
in cells from allergic individuals and inhibit IgE synthesis in vitro. Eur. J. Im-
munol. 29: 2344 –2353.
19. Bohle, B., L. Orel, D. Kraft, and C. Ebner. 2001. Oligodeoxynucleotides con-
taining CpG motifs induce low levels of TNF-
in human B lymphocytes: pos-
sible adjuvants for Th1 responses. J. Immunol. 166: 3743–3748.
20. Horner, A. A., G. F. Widhopf, J. A. Burger, K. Takabayashi, N. Cinman,
A. Ronaghy, H. L. Spiegelberg, and E. Raz. 2001. Immunostimulatory DNA
inhibits IL-4-dependent IgE synthesis by human B cells. J. Allergy Clin. Immu-
nol. 108: 417– 423.
21. Marshall, J. D., S. Abtahi, J. J. Eiden, S. Tuck, R. Milley, F. Haycock, M. J. Reid,
A. Kagey-Sobotka, P. S. Creticos, L. M. Lichtenstein, and G. Van Nest. 2001.
Immunostimulatory sequence DNA linked to the Amb a 1 allergen promotes T
H
1
cytokine expression while down-regulating T
H
2 cytokine expression in PBMCs
from human patients with ragweed allergy. J. Allergy Clin. Immunol. 108:
191–197.
22. Tighe, H., K. Takabayashi, D. Schwartz, G. Van Nest, S. Tuck, J. J. Eiden,
A. Kagey-Sobotka, P. S. Creticos, L. M. Lichtenstein, H. L. Spiegelberg, and
E. Raz. 2000. Conjugation of immunostimulatory DNA to the short ragweed
allergen Amb a 1 enhances its immunogenicity and reduces its allergenicity.
J. Allergy Clin. Immunol. 106: 124 –134.
23. Horner, A. A., K. Takabayashi, L. Beck, B. Sharma, J. Zubeldia, S. Baird,
S. Tuck, L. Libet, H. L. Spiegelberg, F. T. Liu, and E. Raz. 2002. Optimized
conjugation ratios lead to allergen immunostimulatory oligodeoxynucleotide con-
jugates with retained immunogenicity and minimal anaphylactogenicity.
J. Allergy Clin. Immunol. 110: 413– 420.
24. Creticos, P. S., J. T. Schroeder, R. G. Hamilton, S. L. Balcer-Whaley,
A. P. Khattignavong, R. Lindblad, H. Li, R. Coffman, V. Seyfert, J. J. Eiden, and
D. Broide. 2006. Immunotherapy with a ragweed-Toll-like receptor 9 agonist
vaccine for allergic rhinitis. N. Engl. J. Med. 355: 1445–1455.
25. Baldrick, P., D. Richardson, S. R. Woroniecki, and B. Lees. 2007. Pollinex((R))
quattro ragweed: safety evaluation of a new allergy vaccine adjuvanted with
monophosphoryl lipid A (MPL((R))) for the treatment of ragweed pollen allergy.
J. Appl. Toxicol. 27: 399 409.
26. McCormack, P. L., and A. J. Wagstaff. 2006. Ultra-short-course seasonal allergy
vaccine (Pollinex Quattro). Drugs 66: 931–938.
27. Messner, P., F. M. Unger, and U. B. Sleytr. 1996. Vaccine development based on
S-layer technology. In Crystalline Bacterial Cell Surface Proteins. U. B. Sleytr,
P. Messner, D. Pum, and M. Sa´ra, eds. R. G. Landes Comp./Academic Press,
Austin, pp. 161–173.
28. Sleytr, U. B., E. M. Egelseer, N. Ilk, D. Pum, and B. Schuster. 2007. S-Layers as
a basic building block in a molecular construction kit. FEBS J. 274: 323–334.
29. Sleytr, U. B., C. Huber, N. Ilk, D. Pum, B. Schuster, and E. M. Egelseer. 2007.
S-layers as a tool kit for nanobiotechnological applications. FEMS Microbiol.
Lett. 267: 131–144.
30. Breitwieser, A., E. M. Egelseer, D. Moll, N. Ilk, C. Hotzy, B. Bohle, C. Ebner,
U. B. Sleytr, and M. Sara. 2002. A recombinant bacterial cell surface (S-layer)-
major birch pollen allergen-fusion protein (rSbsC/Bet v1) maintains the ability to
self-assemble into regularly structured monomolecular lattices and the function-
ality of the allergen. Protein Eng. 15: 243–249.
31. Bohle, B., A. Breitwieser, B. Zwolfer, B. Jahn-Schmid, M. Sara, U. B. Sleytr, and
C. Ebner. 2004. A novel approach to specific allergy treatment: the recombinant
fusion protein of a bacterial cell surface (S-layer) protein and the major birch
pollen allergen Bet v 1 (rSbsC-Bet v 1) combines reduced allergenicity with
immunomodulating capacity. J. Immunol. 172: 6642– 6648.
32. Ausiello, C. M., M. Cerquetti, G. Fedele, F. Spensieri, R. Palazzo, M. Nasso,
S. Frezza, and P. Mastrantonio. 2006. Surface layer proteins from Clostridium
difficile induce inflammatory and regulatory cytokines in human monocytes and
dendritic cells. Microbes Infect. 8: 2640 –2646.
33. Maurer, D., C. Ebner, B. Reininger, E. Fiebiger, D. Kraft, J. P. Kinet, and
G. Stingl. 1995. The high affinity IgE receptor (FcRI) mediates IgE-dependent
allergen presentation. J. Immunol. 154: 6285– 6290.
34. Jahn-Schmid, B., U. Siemann, A. Zenker, B. Bohle, P. Messner, F. M. Unger,
U. B. Sleytr, O. Scheiner, D. Kraft, and C. Ebner. 1997. Bet v 1, the major birch
pollen allergen, conjugated to crystalline bacterial cell surface proteins, expands
allergen-specific T cells of the Th1/Th0 phenotype in vitro by induction of IL-12.
Int. Immunol. 9: 1867–1874.
35. Netea, M. G., R. Sutmuller, C. Hermann, C. A. Van der Graaf,
J. W. Van der Meer, J. H. van Krieken, T. Hartung, G. Adema, and B. J. Kullberg.
2004. Toll-like receptor 2 suppresses immunity against Candida albicans through
induction of IL-10 and regulatory T cells. J. Immunol. 172: 3712–3718.
36. Hoarau, C., C. Lagaraine, L. Martin, F. Velge-Roussel, and Y. Lebranchu. 2006.
Supernatant of Bifidobacterium breve induces dendritic cell maturation, activa-
tion, and survival through a Toll-like receptor 2 pathway. J. Allergy Clin. Im-
munol. 117: 696 –702.
37. Sato, K., N. Yamashita, M. Baba, and T. Matsuyama. 2003. Modified myeloid
dendritic cells act as regulatory dendritic cells to induce anergic and regulatory T
cells. Blood 101: 3581–3589.
38. Jonuleit, H., E. Schmitt, G. Schuler, J. Knop, and A. H. Enk. 2000. Induction of
interleukin 10-producing, nonproliferating CD4
T cells with regulatory prop-
erties by repetitive stimulation with allogeneic immature human dendritic cells.
J. Exp. Med. 192: 1213–1222.
39. Rutella, S., S. Danese, and G. Leone. 2006. Tolerogenic dendritic cells: cytokine
modulation comes of age. Blood 108: 1435–1440.
7275The Journal of Immunology
... Retained ability of the fusion protein to self-assemble with correctly folded Bet v 1 conformation, but reduced capacity to induce histamine release from blood basophiles, increased IFN-γ-and IL-10-, but no detectable IL-5-secretion from PBMCs isolated from birch pollen-allergic individuals Gerstmayr et al. (2007) Bet v 1 (birch pollen) ...
... Further work on rSbsC-Bet v 1 was published by Gerstmayr et al. (2007). The rSbsC-Bet v 1 fusion protein was shown to preserve Bet v 1-T-cell epitopes, and the potency to induce IFN-γ and IL-10 production in Bet v 1-specific Th2biased T-cell clones while displaying the capacity to activate basophils in mediator-release assays (Gerstmayr et al. 2007). ...
... Further work on rSbsC-Bet v 1 was published by Gerstmayr et al. (2007). The rSbsC-Bet v 1 fusion protein was shown to preserve Bet v 1-T-cell epitopes, and the potency to induce IFN-γ and IL-10 production in Bet v 1-specific Th2biased T-cell clones while displaying the capacity to activate basophils in mediator-release assays (Gerstmayr et al. 2007). Human monocyte-derived DC preparations stimulated with SbsC-Bet v 1 were shown to have a semi-mature phenotype characterized by enhanced surface expression of CD40, CD80, CD83, and CD86 (compared to stimulation with Bet v 1 alone) and a strongly increased secretion of the DC-derived cytokines IL-10 and IL-12 (Gerstmayr et al. 2007). ...
Article
While acute allergic symptoms can be managed by emergency medication, to date, allergen-specific immunotherapy (SIT) with allergen extracts is the only available curative treatment option. However, the risk of anaphylactic reactions, long treatment duration, varying extract quality, and underrepresentation of certain allergens currently prevent many patients from successfully undergoing SIT. Novel strategies are needed to enhance efficacy, safety, and convenience of allergy treatment. Fusion proteins combining allergen and adjuvant into a single molecule can efficiently induce immune responses by targeting the allergen to the relevant immune cells in vivo. Simultaneous co-delivery of both antigen and adjuvant to the same cell in a fixed molecular ratio triggers the uptake and presentation of the conjugated allergen in the context of the adjuvant-induced immune cell activation. This review summarizes the published strategies to improve the treatment of type I allergies using fusion proteins consisting of allergen (peptides) and either (1) immune-activating bacterial (flagellin, MPLA, S-layer, cholera-, and tetanus toxin), (2) viral (PreS, VP-1, TAT), or (3) fungal (FIP-fve) components, (4) immune-activating DNA motifs, (5) forced delivery of allergens to the MHC-II loading pathway, and (6) killing of immune cells expressing allergen-specific IgE by fusion of the allergen to diphtheria toxin.
... Among others, E. coli 083 (57), Helicobacter pylori (83), Clamydia (27), Listeria monocytogenes (85), Mycobacterium vaccae (111), and Bacillus Calmette-Guérin (BCG) (84) were reported to induce DC-derived IL-10 secretion with immune modulatory potential for the treatment of allergic diseases. Of note, in contrast to IL-10-treated DCs, many of the available studies suggest that stimulation with either bacteria (live or killed) or isolated bacterial components induces both anti-inflammatory IL-10-and pro-inflammatory, Th1-promoting IL-12 secretion often alongside an enhanced expression of co-stimulatory molecules on the stimulated DCs ( Figure 3A) (27,84,86,98,112). ...
... Moreover, Gerstmayr and colleagues generated a recombinant fusion protein of a bacterial surface (S-layer) protein of Geobacillus stearothermophilus (SbsC) and the major birch pollen allergen Bet v 1 as a vaccine candidate to improve the treatment of birch pollen allergy. The SbsC:Bet v 1 fusion protein displayed reduced mediator-releasing capacity, while both preserving Bet v 1-T cell epitopes, and the potency to induce IFN-γ and IL-10 production in Bet v 1-specific Th2-biased T cell clones (98). DCs stimulated with the fusion protein were shown to have a semi-mature phenotype characterized by enhanced expression of CD40, CD80, and CD86 which were still lower than the levels induced by stimulation with LPS ( Figure 3A) (98). ...
... The SbsC:Bet v 1 fusion protein displayed reduced mediator-releasing capacity, while both preserving Bet v 1-T cell epitopes, and the potency to induce IFN-γ and IL-10 production in Bet v 1-specific Th2-biased T cell clones (98). DCs stimulated with the fusion protein were shown to have a semi-mature phenotype characterized by enhanced expression of CD40, CD80, and CD86 which were still lower than the levels induced by stimulation with LPS ( Figure 3A) (98). Moreover, the SbsC:Bet v 1 fusion protein strongly increased DC-derived IL-10 and IL-12 secretion (98). ...
Article
Full-text available
Dendritic cells (DCs) are gatekeepers of the immune system that control induction and polarization of primary, antigen-specific immune responses. Depending on their maturation/activation status, the molecules expressed on their surface, and the cytokines produced DCs have been shown to either elicit immune responses through activation of effector T cells or induce tolerance through induction of either T cell anergy, regulatory T cells, or production of regulatory cytokines. Among the cytokines produced by tolerogenic DCs, interleukin 10 (IL-10) is a key regulatory cytokine limiting und ultimately terminating excessive T-cell responses to microbial pathogens to prevent chronic inflammation and tissue damage. Because of their important role in preventing autoimmune diseases, transplant rejection, allergic reactions, or in controlling chronic inflammation DCs have become an interesting tool to modulate antigen-specific immune responses. For the treatment of allergic inflammation, the aim is to downregulate allergen-specific T helper 2 (Th2) responses and the associated clinical symptoms [allergen-driven Th2 activation, Th2-driven immunoglobulin E (IgE) production, IgE-mediated mast cell and basophil activation, allergic inflammation]. Here, combining the presentation of allergens by DCs with a pro-tolerogenic, IL-10-producing phenotype is of special interest to modulate allergen-specific immune responses in the treatment of allergic diseases. This review discusses the reported strategies to induce DC-derived IL-10 secretion for the suppression of allergen-specific Th2-responses with a focus on IL-10 treatment, IL-10 transduction, and the usage of both whole bacteria and bacteria-derived components. Interestingly, while IL-10-producing DCs induced either by IL-10 treatment or IL-10 transduction are arrested in an immature/semi-mature state, treatment of DCs with live or killed bacteria as well as isolated bacterial components results in the induction of both anti-inflammatory IL-10 and pro-inflammatory, Th1-promoting IL-12 secretion often paralleled by an enhanced expression of co-stimulatory molecules on the stimulated DCs. By the secretion of DC-derived exosomes or CC-chemokine ligand 18, as well as the expression of inhibitory molecules like cytotoxic T lymphocyte-associated antigen 4, TNF receptor superfamily member 4, Ig-like transcript-22/cluster of differentiation 85, or programmed death-1, IL-10-producing DCs have been repeatedly shown to suppress antigen-specific Th2-responses. Therefore, DC-based vaccination approaches hold great potential to improve the treatment of allergic diseases.
... However, the issue of inefficiency of AIT with individual allergens has not been resolved. Therefore, combining allergens with TLR agonists is an appealing solution for improving efficacy and long-term tolerance [1], such as recombinant fusion proteins with bacterial surface (S-) layer proteins that activate TLR2 receptor [41] and TLR5 agonist flagellin [42] have been developed, as well as chemically conjugated allergens to detoxified TLR4-ligand MPLA [43], or immunostimulatory oligodeoxynucleotides (ISS-ODN) containing TLR9activating unmethylated CpG motifs [44]. The sublingual administration of the vaccine in AIT is attractive due to its safety and convenient self-administration and is recommended by the World Health Organization-Allergic Rhinitis and its Impact on Asthma (ARIA) [45]. ...
Article
Full-text available
Novel allergen immunotherapy (AIT) approaches necessitate the use of more effective and safe therapeutics, which can be accomplished by employing novel adjuvants for improved innate immune cell activation, as well as hypoallergenic allergen forms. In this study, we investigate the immunomodulatory effects of a chimera rBet v 1a-BanLec wt (rBv1a-BLwt; C wt) composed of the major birch pollen allergen Bet v 1a and banana lectin (BanLecwt; BLwt) and two novel chimeras, rBv1l-BL H84T (rBet v 1l-BanLec H84T ; C1) and rBL H84T-Bv1l (rBanLec H84T-Bet v 1l; C2), both composed of BL H84T and hypoallergenic birch pollen allergen Bv1l in the co-culture model Caco-2/ THP-1, and PBMCs from donors with birch pollen allergy. The chimeric molecules rBv1l-BL H84T (C1) and rBLH84T-Bv1l (C2) were created in silico and then produced in E. coli using recombinant DNA technology. Real-time PCR analysis of gene expression following compound treatment in the co-culture model revealed that all three chimeras have the potential to induce the anti-inflammatory cytokine IL-10 gene expression in Caco-2 cells and IFN-γ gene expression in THP-1 cells. Sandwich ELISA revealed that Cwt increased IL-10 secretion and IFN-/ IL-4 levels in PBMCs from birch pollen allergic donors, whereas C1 and C2 were less effective. The findings suggest that Cwt should be analyzed further due to its potential benefit in AIT.
... Conjugating recombinant allergens with SLPs leads to strongly reduced IgE-binding activity and promotes the induction of allergen-specific Th0/1 cells and regulatory T cells. This type of allergen modification has been attempted with inhalant allergens (83). Subsequently, bacterial S-layers have been studied as carriers for peanut allergen-derived peptides (84,85). ...
Article
Full-text available
Allergen immunotherapy is currently the only causal treatment for allergic diseases in human beings and animals. It aims to re-direct the immune system into a tolerogenic or desensitized state. Requirements include clinical efficacy, safety, and schedules optimizing patient or owner compliance. To achieve these goals, specific allergens can be formulated with adjuvants that prolong tissue deposition and support uptake by antigen presenting cells, and/or provide a beneficial immunomodulatory action. Here, we depict adjuvant formulations being investigated for human and veterinary allergen immunotherapy.
... In the following years, recombinant fusion of the Bet v 1 to S-layer proteins successfully replaced the procedures of chemical coupling. For example, recombinant fusion of Bet v 1 with S-layer proteins, SbpA from Bacillus sphaericus and SbsC from Bacillus stearothermophilus, yielded two S-layer/allergen recombinant constructs, which showed strongly reduced capacity to bind IgE compared to free Bet v 1 and possessed the ability to induce allergen-specific T H 0/T H 1 and regulatory T cell immune responses Ilk et al., 2002;Gerstmayr et al., 2007). Initially, the S-layer/allergen fusion proteins were expressed in Gram-negative host E. coli and the associated endotoxin was subsequently removed by purification procedure, which is costly and time-consuming. ...
Article
Full-text available
Allergen-specific immunotherapy represents the only available curative approach to allergic diseases. The treatment has proven effective, but it requires repetitive administrations of allergen extracts over 3–5 years and is often associated with adverse events. This implies the need for novel therapeutic strategies with reduced side effects and decreased treatment time, which would improve patients’ compliance. Development of vaccines that are molecularly well defined and have improved safety profile in comparison to whole allergen extracts represents a promising approach. Molecular allergy vaccines are based on major allergen proteins or allergen-derived peptides. Often, such vaccines are associated with lower immunogenicity and stability and therefore require an appropriate delivery vehicle. In this respect, viruses, bacteria, and their protein components have been intensively studied for their adjuvant capacity. This article provides an overview of the microbial delivery vehicles that have been tested for use in allergy immunotherapy. We review in vitro and in vivo data on the immunomodulatory capacity of different microbial vehicles for allergens and allergen-derived peptides and evaluate their potential in development of allergy vaccines. We also discuss relevant aspects and challenges concerning the use of microbes and their components in immunotherapy of allergic diseases.
... Intracellular cytokine staining showed that rSbsC-Bet-v1 promoted Th cells production by IFNγ. Furthermore, rSbsC-Bet-v-1 induced IFNγ synthesis in Bet-v1-specific Th2 cells, and the more important is an increase in IL-10 production in these cells (Bohle et al., 2004;Gerstmayr et al., 2007). ...
Article
In recent years, increasing attention has been paid to bioengineered bacteria as vectors for the treatment of allergic diseases. The methods for preparing bioengineered bacteria that can express exogenous genes are improving. Research has focused mainly on application of bioengineered bacteria expressing recombinant allergens, hypoallergenic derivatives of allergens, T-cell epitope derivatives, cytokines, or as mucosal adjuvants to enhance immunotherapy effects. This strategy offers new ideas for the treatment of allergic diseases. This review summarizes recent advances in use of live bioengineered bacteria in allergic diseases as well as the challenges of using microorganisms (or their components) in immunotherapy.
Article
Allergen immunotherapy (AIT) is the only disease-modifying treatment option for patients with type 1–mediated allergic diseases such as allergic rhinitis/rhinoconjunctivitis with/without allergic asthma. Although many innovations have been developed since the first clinical report of Noon et al in 1911, the improvement of clinical efficacy and tolerability of this treatment is still an important unmet need. Hence, much progress has been made in the characterization of the cell types, cytokines, and intracellular signaling events involved in the development, maintenance, and regulation of allergic reactions, and also in the understanding of the mechanisms of tolerance induction in AIT. This comprehensive review aims to summarize the current innovative approaches in AIT, but also gives an outlook on promising candidates of the future. On the basis of an extensive literature review, integrating a clinical point of view, this article focuses on recent and future innovations regarding biologicals, allergen-derived peptides, recombinant allergens, “Toll”-like receptor agonists and other adjuvants, and novel application routes being developed for future AIT.
Article
Certain naturally occurring proteins consist of a number of subunit building blocks that are capable of self-assembling to form nanoscale particles with highly organized, symmetrical, and homogeneous structures. These protein- based nanoparticles have high surface/volume ratios and other favorable properties, including mono-dispersibility, high stability, low toxicity, biocompatibility, biodegradability, and capacity for easy genetic and chemical modification. Thus, these particles have attracted considerable research attention and have been manipulated for various applications in different fields. This review describes the engineering of existing protein nanoparticles, with a particular focus on scientific advances in diverse applications, including bioassays, molecular imaging diagnostics, drug delivery, biocatalysis, and materials science. In addition, barriers for the widespread industrial use of such nanoparticles and outlook for the design and creation of novel self-assembled protein-based nanoparticles are considered.
Article
Full-text available
Commensal gut bacteria are essential for the development and maintenance of the gut's immune system. Some bacteria strains, such as Lactobacillus and Bifidobacterium species, have been reported to provide protection from allergic and inflammatory bowel diseases. However, the interactions between these commensal bacteria and the immune system are largely unknown. Objective: We studied the effects of a supernatant from the culture of B breve C50 (BbC50) on the maturation, activation, and survival of human dendritic cells (DCs). Methods: DCs were differentiated from human monocytes with IL-4 and GM-CSF for 5 days and cultured with BbC50 supernatant (BbC50 SN) or LPS for 2 days. Results: BbC50 SN induced DC maturation, with increase in CD83, CD86, and HLA-DR expression. We also showed, for the first time, that BbC50 SN prolonged DC survival, with high IL-10 and low IL-12 production compared with that seen in LPS-DCs. Moreover, BbC50 SN inhibited the effects of LPS on DCs, both in terms of IL-12 production and in terms of survival. The prolonged DC survival was independent of IL-10 production and nuclear factor kB pathway but was associated with an upregulation of Bcl-x L and Phospho-Bad. Finally, BbC50 SN induced activation of Toll-like receptor 2 (TLR2)–transfected cells in contrast to TLR4-, TLR7-, and TLR9-transfected cells. Conclusion: The supernatant of B breve C50 can induce DC maturation and prolonged DC survival through TLR2, with high IL-10 production. These properties might correspond to a regulatory DC profile, which could limit the excessive T H 1 response and control the excessive T H 2 polarization observed in atopic newborns. (J Allergy Clin Immunol 2006;117:696-702.) Live commensal microflora is essential for the development of the gut's immune system. 1 Some bacteria, such as Lactobacillus and Bifidobacterium species, described as ''living microorganisms exerting health benefit,'' 2 define the concept of probiotics. These bacteria have been reported to prevent and treat rotavirus infections and postantibiotic diarrhea, 3,4 allergic diseases, 5-7 and recurrence of inflammatory bowel disease. 8,9 These studies suggest that the intestinal immune system could be a privileged target of probiotics. Nevertheless, despite several studies reporting modifications of immunologic parameters induced by probiotic bacteria, the interactions between the cells of the intestinal immune system and bacteria remain largely unknown. 10 However, it has been shown that killed probiotic bacteria can affect the maturation and cytokine secretion profile of dendritic cells (DCs), 11,12 which represent potent antigen-presenting cells. DCs also have properties to induce negative regulation , with generation of regulatory T cells. 13 DCs can be activated by bacterial components through the interaction between pathogen-derived immunostimulatory molecules from bacteria and membrane receptors called pattern-recognition receptors, including the Toll-like receptor (TLR) family. 14 The different TLRs recognize a broad spectrum of highly conserved microbial structures, such as proteins, lipids, glycoproteins, and nucleic acid motifs, contained in the wall, cytoplasm, and nucleus of bacteria. TLR engagement could therefore have different types of effects on DC activation according to the bacteria strain. 15 Because of these properties, DCs represent a potential target of probi-otic bacteria. Menard et al 16 recently reported that active bacterial products from Bifidobacterium breve C50 (BbC50) could cross an intestinal monolayer of epithelial cells. We therefore studied the effects of a supernatant of BbC50 (BbC50 SN) on human monocyte-derived DCs in vitro to define the interactions with the immune system. In our model BbC50 SN was able to mature and fully activate DCs, inducing a particular cytokine synthesis profile and prolonged DC survival through a TLR2 pathway. Abbreviations used 7-AAD: 7-Amino actinomycin D BbC50: Bifidobacterium breve C50 BbC50 SN : Supernatant of Bifidobacterium breve C50 BbC50 SN-DC: DC treated with BbC50 SN DC: Dendritic cell NF: Nuclear factor PE: Phycoerythrin TLR: Toll-like receptor
Article
Full-text available
The effects of phosphorothioate oligonucleotides containing CpG motifs (CpG-ODN) on cultured cells from allergic patients and non-atopic individuals were investigated. In peripheral blood mononuclear cells (PBMC) CpG-ODN led to a significant increase of IFN-γ. By intracellular cytokine staining, IFN-γ production could be attributed to NK cells and inhibition experiments indicated an IL-12-dependent mechanism. Moreover, CpG-ODN increased mRNA expression of IL-12 and IL-18 in PBMC. In this respect, no significant difference between allergic and non-atopic individuals was observed. Monocyte-derived dendritic cells were identified as one IL-12- and IL-18-producing source. In addition, stimulation of PBMC derived from atopic patients with CpG-ODN led to a considerable increase of polyclonal IgG and IgM synthesis. In contrast, the production of total IgE was suppressed. CpG-ODN induced a significant rise of IgG and IgM specific for allergens to which the patients were sensitized, whereas allergen-specific IgE levels remained unchanged. Our data suggest that CpG-ODN display a strong influence on the ongoing immune response and might represent potential adjuvants for specific immunotherapy of type I allergy.
Article
Full-text available
The discovery that the high affinity IgE receptor (Fc epsilon RI) is expressed on APCs of patients with atopic diseases raised the possibility that the functional importance of Fc epsilon RI in the pathogenesis of atopy may extend beyond its role in type I allergic reactions. Here we show that, following removal of in vivo-bound IgE by lactic acid treatment, targeting of allergens to monocytes by Ag-specific IgE critically depends on Fc epsilon RI expression. Even more importantly, lactic acid-treated, monocyte-enriched PBMCs present allergen to T cells 100- to 1000-fold more effectively if the allergen has been targeted to Fc epsilon RI on these cells via allergen-specific IgE. This mechanism may critically lower the atopic individual's threshold to mount allergen-specific T cell responses capable of promoting IgE production and delayed-type hypersensitivity reactions.
Article
Full-text available
Allergen specific CD4+ T cell clones generated from allergic individuals have been shown to produce increased levels of the cytokine interleukin 4 (IL-4), compared to allergen specific clones generated from nonallergic individuals. This difference between CD4+ T cells from allergic and nonallergic individuals with regard to cytokine production in response to allergen is thought to be responsible for the development of allergic disease with increased IgE synthesis in atopic individuals. We examined the production of IL-4 in subjects with allergic rhinitis and in allergic individuals treated with allergen immunotherapy, a treatment which involves the subcutaneous administration of increasing doses of allergen and which is highly effective and beneficial for individuals with severe allergic rhinitis. We demonstrated that the quantity of IL-4 produced by allergen specific memory CD4+ T cells from allergic individuals could be considerably reduced by in vivo treatment with allergen (allergen immunotherapy). Immunotherapy reduced IL-4 production by allergen specific CD4+ T cells to levels observed with T cells from nonallergic subjects, or to levels induced with nonallergic antigens such as tetanus toxoid. In most cases the levels of IL-4 produced were inversely related to the length of time on immunotherapy. These observations indicate that immunotherapy accomplishes its clinical effects by reducing IL-4 synthesis in allergen specific CD4+ T cells. In addition, these observations indicate that the cytokine profiles of memory CD4+ T cells can indeed be altered by in vivo therapies. Thus, the cytokine profiles of memory CD4+ T cells are mutable, and are not fixed as had been suggested by studies of murine CD4+ memory T cells. Finally, treatment of allergic diseases with allergen immunotherapy may be a model for other diseases which may require therapies that alter inappropriate cytokine profiles of memory CD4+ T cells.
Article
Full-text available
Modulation of allergic immune responses by using adequate adjuvants is a promising concept for future immunotherapy of type I hypersensitivity. In the present study, recombinant Bet v 1 (rBet v 1, the major birch pollen allergen) was conjugated to cross-linked crystalline surface layer proteins (SL) derived from Gram-positive eubacteria. T cell lines (TCL) and clones (TCC) were established from peripheral blood of birch pollen-allergic patients. TCL and TCC were induced either using rBet v 1 alone or rBet v 1/SL conjugates (rBet v 1/SL) as initial antigen stimulus. Cytokine production after re-stimulation with rBet v 1 was investigated. TCL initiated with rBet v 1/SL showed significantly increased IFN-gamma production as compared to rBet v 1 -selected TCL. TCC were established from TCL of five patients. As expected, the majority of CD4+ TCC induced by rBet v 1 (55%) displayed a Th2-like pattern of cytokine production. However, only 21% of Bet v 1-specific TCC isolated from TCL established with the Bet v 1/SL revealed this phenotype. The majority of SL-specific TCC (80%) belonged to the Th1 phenotype. In cultures of peripheral blood mononuclear cells, both, SL and Bet v 1/SL (but not rBet v 1) stimulated the production of high levels of IL-12, a pivotal mediator of Th1 responses. Moreover, stimulation of rBet v 1-induced TCC with rBet v 1/SL led to an increased IFN-gamma production. This effect could be reversed by neutralizing anti-IL-12 mAb. Together these results indicate an adjuvant effect of SL mediated by IL-12. Our results indicate that bacterial components, such as SL, displaying adjuvant effects may be suitable for immunotherapeutical vaccines for type I allergy.
Article
Full-text available
The induction of allergen-specific anergy in peripheral T cells represents a key step in specific immunotherapy (SIT). Here we demonstrate that the anergic state results from increased IL-10 production. In bee venom (BV)-SIT the specific proliferative and cytokine responses against the main allergen, the phospholipase A2 (PLA), and T cell epitope-containing PLA peptides were significantly suppressed after 7 d of treatment. Simultaneously, the production of IL-10 increased during BV-SIT. After 28 d of BV-SIT the anergic state was established. Intracytoplasmic cytokine staining of PBMC combined with surface marker detection revealed that IL-10 was produced initially by activated CD4(+)CD25(+), allergen-specific T cells, and followed by B cells and monocytes. Neutralization of IL-10 in PBMC fully reconstituted the specific proliferative and cytokine responses. A similar state of IL-10-associated T cell anergy, as induced in BV-SIT, was found in hyperimmune individuals who recently had received multiple bee stings. The addition of IL-10 to soluble CD40 ligand IL-4-stimulated PBMC or purified B cells inhibited the PLA-specific and total IgE and enhanced the IgG4 formation. Accordingly, increased IL-10 production by SIT causes specific anergy in peripheral T cells, and regulates specific IgE and IgG4 production toward normal IgG4-related immunity.
Chapter
D uring the past rwo decades a considerable body of know­ ledge has been assembled on ultrastructure, chemistry, and molecular genetics of S-layers 1. 7 (see also chapters 2 to 4 of this volume). Bacterial S-layers have been successfully used for the de­ velopment of isoporous ultrafiltration membranes, as afflnity matrices, as supports for Langmuir-Blodgett films and biological membranes, for the production of S-layer-stabilized liposomes, in molecular nanotechnologyS·9 (see also chapters 6 and 8 of this volume) and as an expression system for biotechnologically useful 12 recombinant proteins. 10. Other imponant applications were the use of S-layers for vaccinating fish 13. 16 and as carrier/adjuvants for vaccination and immunotherapy.17.19 7.2. S-LAYERS AS A FISH VACCINE Fish represent a major source of food in the world. The in­ creasing world population will continue to demand more fish protein in the future because it is tasty, wholesome and relatively inexpensive to produce. Therefore, fish culture is of increasing importance. Like other animals, fish are susceptible to microbial disease, especially when subjected to intensive culture practices. 14 Crys1alline Baccerial Cell Surface Proceins, edited by Uwe B. Sleytr, Paul Messner, Dietmar Pum, Margit Sara .
Article
The mechanisms of bee venom immunotherapy (VIT) are largely unknown. The aim of this study was to follow the changes of T cell cytokine secretion during the course of VIT. Ten bee venom-allergic patients with a history of severe systemic reactions, positive skin tests, and bee venom (BV)-specific serum IgE Abs were treated as follows: on the first day, a cumulative dose of 111 micrograms, starting with 0.1 microgram, was administered s.c. under intensive care conditions. Further injections of 100 micrograms BV were given on day 7, day 21, and thereafter at intervals of 4 wk. Blood samples were obtained just before the initiation of VIT, after the last injection on the same day, and before the subsequent BV injections on days 7, 21, and 50 of VIT. Peripheral blood mononuclear cells (PBMC) were stimulated with phospholipase A (PLA), the major BV allergen, or with a control Ag tetanus toxoid (TT). Cytokine secretion was measured 24 h after restimulation of the cultures with solid-phase bound OKT3 F(ab')2 mAbs after 7 days of culture. In PLA-stimulated cultures, VIT resulted in decreased IL-4 and IL-5 and increased IFN-gamma secretion. In TT-stimulated cultures, we observed similar levels of cytokines before and during VIT. We conclude that ultra-rush VIT changes allergen-specific T cell reactivity.
Article
Grass pollen injection immunotherapy is effective in patients with summer hay fever, although efficacy must be balanced against possible side effects. The mechanism of immunotherapy is unknown but may be related to its ability to inhibit allergen-induced late responses, which are known to be characterized by infiltration of T lymphocytes, eosinophils, and cells with messenger RNA for so-called TH2-type cytokines (IL-4 and IL-5). This study was designed to observe the effect of grass pollen immunotherapy on late nasal responses and associated cellular infiltration and cytokine mRNA expression. We performed local nasal provocation with grass pollen (and a control challenge) in 28 patients after a 12-month double-blind, placebo-controlled trial of immunotherapy. Nasal biopsy specimens were obtained at 24 hours and processed for immunohistology and in situ hybridization studies. Grass pollen immunotherapy inhibited allergen-induced immediate (0 to 60 minutes) increases in sneezing (p < 0.02) and nasal blocking (p < 0.01) and late (0 to 24 hours) nasal symptoms (p < 0.05). Immunotherapy also inhibited the associated infiltration of the nasal mucosa by CD4+ T lymphocytes and total (major basic protein-containing) and "activated" (cationic protein-secreting) eosinophils (all p = 0.03). There was a significant (p = 0.04) increase in cells expressing mRNA for interferon-gamma at 24 hours after allergen challenge, which correlated inversely with patients' seasonal symptoms (r = -0.65, p < 0.05) and medication requirements (r = -0.75, p < 0.02) during the pollen season. The results suggest that successful grass pollen immunotherapy for summer hay fever may act by inhibiting allergen-induced T lymphocyte and eosinophil recruitment and eosinophil activation in the target organ, possibly through a mechanism involving protective local increases in TH1-type cells.
Article
The current study was carried out to elucidate the immunoregulatory changes induced by venom immunotherapy (VIT) in bee or wasp allergic subjects. All subjects included in this study had a history of severe systemic allergic reactions to stings of the respective insect as well as positive skin tests with the respective venom or venom-specific IgE in the sera. Parameters assessed in peripheral blood mononuclear cells (PBMC) before and after initiation of VIT (rush therapy reaching a maintenance dose of 100 micrograms venom injected subcutaneously within 1 week) were expression of CD3, CD4, CD8, CD45RA, CD45RO, interleukin (IL)-2 receptor (R) alpha, IL-4R, IL-12R, Fc epsilon RII, CD40, and CD40 ligand (CD40L), cells producing interferon (IFN)-gamma and IL-10 after stimulation with phorbol 12-myristate 13-acetate + ionomycin in the presence of monensin measured by flow cytometry; secretion of IFN-gamma, IL-4, and IL-10 measured by ELISA (IFN-gamma and IL-10 were additionally measured by PCR), and proliferation after stimulation with the respective venom. Significant decreases were observed after VIT for proliferative response to venom and venom + IL-4, IL-4 secretion, Fc epsilon RII, CD40, and CD40L expression. Significant increases were observed after VIT for IFN-gamma concerning the amount secreted and the number of producing cells, and IL-10, IL-10 was mainly produced by CD4+ cells that were negative for IFN-gamma, but some double-positive (IL-10 and IFN-gamma) cells were always detected. Addition of blocking anti-IL-10 antibodies, but not isotype control antibodies, prevented down-regulation of proliferation (but not IL-4 secretion) and further enhanced IFN-gamma secretion after VIT. These data indicate that in insect venom allergic subjects, VIT not only induces a rapid shift in cytokine expression from Th2 to Th1 cytokines, but also leads to induction of the immunosuppressive cytokine IL-10, which may be important for the limitation of potentially harmful allergen-specific Th1 responses. The described changes in cytokine expression may be responsible for subsequent increases in allergen-specific IgG and decreases in IgE production, as well as suppressive activity observed in earlier studies.