The Wnt/b-Catenin Pathway Attenuates Experimental Allergic

Transcrição

The Wnt/b-Catenin Pathway Attenuates Experimental Allergic
The Wnt/β-Catenin Pathway Attenuates
Experimental Allergic Airway Disease
This information is current as
of January 20, 2017.
Sebastian Reuter, Helen Martin, Hendrik Beckert, Matthias
Bros, Evelyn Montermann, Christina Belz, Anke Heinz,
Svetlana Ohngemach, Ugur Sahin, Michael Stassen, Roland
Buhl, Leonid Eshkind and Christian Taube
J Immunol published online 13 June 2014
http://www.jimmunol.org/content/early/2014/06/13/jimmun
ol.1400013
http://www.jimmunol.org/content/suppl/2014/06/13/jimmunol.140001
3.DCSupplemental.html
Subscriptions
Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscriptions
Permissions
Email Alerts
Submit copyright permission requests at:
http://www.aai.org/ji/copyright.html
Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/cgi/alerts/etoc
The Journal of Immunology is published twice each month by
The American Association of Immunologists, Inc.,
9650 Rockville Pike, Bethesda, MD 20814-3994.
Copyright © 2014 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
Supplementary
Material
Published June 13, 2014, doi:10.4049/jimmunol.1400013
The Journal of Immunology
The Wnt/b-Catenin Pathway Attenuates Experimental
Allergic Airway Disease
Sebastian Reuter,* Helen Martin,* Hendrik Beckert,* Matthias Bros,† Evelyn Montermann,†
Christina Belz,* Anke Heinz,* Svetlana Ohngemach,‡ Ugur Sahin,x Michael Stassen,{
Roland Buhl,* Leonid Eshkind,‡,1 and Christian Taube‖,1
O
ver the last few decades, the incidence and prevalence of
asthma, especially in Western countries, have increased
dramatically. In recent years, it has become clear that
asthma is a heterogeneous disorder based on multiple pathophysiological mechanisms that are thought to contribute to the different
phenotypes of the disease (1). One prominent phenotype is allergic
asthma, in which Th2 cells and Th2 cytokines, such as IL-4, IL-5, and
IL-13, play a central role (2, 3). The mechanisms leading to asthma
are affected by various interacting parameters, such as genetic predisposition, environmental factors, and exposure to allergens and
*Pulmonary Department, III. Medical Clinic, University Medical Center, Johannes
Gutenberg University Mainz, 55131 Mainz, Germany; †Department of Dermatology,
University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz,
Germany; ‡Transgenic Facility, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; xIII. Medical Clinic, University Medical
Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; {Institute
of Immunology, University Medical Center, Johannes Gutenberg University Mainz,
55131 Mainz, Germany; and ‖Department of Pulmonology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
1
L.E. and C.T. contributed equally to this work.
Received for publication January 3, 2014. Accepted for publication May 8, 2014.
This work was supported by the University Medical Center, Johannes Gutenberg
University Mainz.
Address correspondence and reprint requests to Dr. Christian Taube or Dr. Sebastian
Reuter, Department of Pulmonology, Leiden University Medical Center, P.O. Box
9600, Albinusdreef 2, Room C3-P, 2300 RC Leiden, The Netherlands (C.T.) or
Pulmonary Department, III. Medical Clinic, University Medical Center, Johannes
Gutenberg University Mainz, Langenbeckstraße 1, Building 302T, Room 01,
55131 Mainz, Germany (S.R.). E-mail addresses: [email protected] (C.T.) or
[email protected] (S.R.)
The online version of this article contains supplemental material.
Abbreviations used in this article: AHR, airway hyperresponsiveness; BALF, bronchoalveolar lavage fluid; BMDC, bone marrow–derived dendritic cell; DC, dendritic
cell; DOX, doxycycline; GSK-3b, glycogen synthase kinase 3beta; i.n., intranasal
(ly); PAS, periodic acid–Schiff; pDC, plasmacytoid dendritic cell; SB, SB216763;
tLN, draining lymph node; TM, test medium; Treg, regulatory T cell.
Copyright Ó 2014 by The American Association of Immunologists, Inc. 0022-1767/14/$16.00
www.jimmunol.org/cgi/doi/10.4049/jimmunol.1400013
infectious organisms. This can lead to sensitization against harmless
Ags, resulting in allergic disease following repeated contact with
the Ag/allergen. Dendritic cells (DCs) are the most effective cells
for Ag presentation and represent sentinels of the immune system.
When harmless self-Ag or environmental Ag is encountered, DCs
induce tolerance or anergy (4, 5). However, in the presence of endogenous or exogenous danger signals, DCs become activated and
induce an Ag-specific adaptive immune reaction, which leads to
a memory response and sensitization. Via a spectrum of different
mediators, DCs determine the type and strength of adaptive immune
responses and play an important role in the development of the
allergic response. Indeed, therapeutic modulation of DCs could be
an efficient step to abrogate allergen-induced immune responses.
Recently, the Wnt/b-catenin pathway has been identified as
important in developmental processes in embryogenesis (6). Wnt
proteins are a group of 19 hydrophobic secreted glycoproteins.
Wnt signals via binding to frizzled receptors that form complexes
with the low-density lipoprotein receptor–related protein 5 or 6.
Depending on the ligand/receptor combination, three signal cascades can be induced. First, activation of the canonical Wnt/
b-catenin pathway prevents glycogen synthase kinase 3beta
(GSK-3b)–mediated b-catenin degradation, leading to nuclear
translocation of b-catenin and gene transcription (7). In contrast,
noncanonical Wnt pathways do not use b-catenin signaling; they
include the planar cell polarity pathway, which contributes to the
regulation of tissue polarity and cell migration (8), and a Ca2+mediated pathway, which inhibits b-catenin signaling and also
induces migration (9). The canonical Wnt/b-catenin pathway is
important for self-renewing processes in the course of development via the control of cell proliferation and terminal differentiation, as demonstrated in the gut (10, 11), hair (12), regulation of
hematopoietic stem cells (13), and homeostasis of bone (14, 15).
Moreover, Wnt ligands were shown to play an important role in
developmental and homeostatic processes in the lung (16). Indeed,
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
Signaling via the Wnt/b-catenin pathway plays crucial roles in embryogenesis and homeostasis of adult tissues. In the lung, the
canonical Wnt/b-catenin pathway has been implicated in remodeling processes, development of emphysema, and fibrosis. However, its relevance for the modulation of allergic responses in the lung remains unclear. Using genetically modified mice with lungspecific inducible (doxycycline) Wnt-1 expression (CCSP-rtTA 3 tetO-Wnt1), the impact of Wnt on the development of allergic
airway disease was analyzed. Overexpression of Wnt during the allergen challenge phase attenuated the development of airway
inflammation in an acute model, as well as in a more therapeutic model of secondary challenge. These findings were further
supported by treatment of allergen-sensitized mice with LiCl during challenge. Similar to Wnt, LiCl prevented the degradation of
b-catenin and, thus, attenuated allergic airway inflammation and hyperresponsiveness. Migration studies revealed that lungspecific expression of Wnt reduced the migration of Ag-loaded dendritic cells (DCs) into the draining lymph nodes following
allergen challenge. Administration of in vitro allergen-loaded DCs overcame Wnt-mediated suppression of airway inflammation.
Furthermore, in vitro studies confirmed that DC-dependent T cell activation is impaired by blocking b-catenin degradation. These
results demonstrate an important role for the canonical Wnt/b-catenin pathway in the DC-mediated regulation of allergic
responses in the lung. The Journal of Immunology, 2014, 193: 000–000.
2
Wnt-7b–deficient mice exhibit hypoplastic lungs (17), and a loss of
Wnt-5a signaling induces increased cell proliferation, leading to
a thickened interstitium and distal bronchial branching (18). Deregulation of Wnt molecules in the adult lung has been linked to formation of tumors (19), emphysema, and lung fibrosis (20, 21). In
contrast, little is known about the involvement of Wnt in inflammatory and immunological responses in the lung, such as allergic airway
inflammation.
Therefore, the aim of the current study was to analyze the
function of Wnt-1/b-catenin signaling in the development and
exacerbation of experimental allergic airway disease. In the current study, we observed that overexpression of Wnt-1 resulted in
an attenuation of allergic airway disease, both in acute and therapeutic models.
Materials and Methods
Mice
Assessment of allergic airway disease
Measurement of airway resistance and dynamic compliance were performed on anesthetized, intubated, and mechanically ventilated mice
(flexiVent; Scireq, Montreal, QC, Canada), as previously described (28).
Lungs were lavaged with 1 ml PBS. Cell count and viability assessment
were determined via trypan blue exclusion. Differential cell counts for
macrophages, lymphocytes, neutrophils, and eosinophils were performed
on cytocentrifuged preparations stained with a Hemacolor Set (Merck).
Bronchoalveolar lavage fluid (BALF) supernatants were stored at 220˚C.
Lungs were fixed by inflation and immersion in 10% formalin and embedded in paraffin, as previously described (28). Tissue sections were
stained with H&E or periodic acid–Schiff (PAS). To assess airway inflammation, five randomly selected areas were scored for each slide by two
experienced observers blinded to the experimental groups (29). Inflammation was scored on a scale from 0 to 4. PAS+ goblet cells were quantified per millimeter of bone marrow.
b-catenin staining was performed as recommended by the manufacturer.
Slides were incubated with primary b-catenin Ab (R&D Systems, Minneapolis, MN) or isotype control in a final concentration of 2.5 mg/ml
overnight at 4˚C. Secondary Ab, as well as HRP conjugate, was incubated on slides, as described in the staining kit (Anti-Goat HRP-DAB;
R&D Systems). Slides were analyzed and scored in a blinded fashion by
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
C57BL/6J, B6 OTII, DO11.10, and BALB/c mice were obtained from the
Central Laboratory Animal Facility, University Medical Center, Johannes
Gutenberg University Mainz. CCSP-rtTA and tetO-Wnt1 mice were generated as described in earlier reports (22, 23). Transgenic mice were
maintained by crosses with wild-type FVB/N animals. CCSP-rtTA–transgenic mice were bred with tetO-Wnt1 animals to generate bitransgenic
mice. Animals were genotyped using tail DNA. All mice used were aged
8–12 wk of age. Animal procedures were conducted in accordance with
current federal, state, and institutional guidelines, and all experiments were
approved by the local regulatory authorities.
Wnt/b-CATENIN ATTENUATES ASTHMA
Experimental protocols
To assess the effects of Wnt-1/b-catenin signaling, different experimental
animal models were carried out as summarized in Fig. 1. For sensitization
to the model Ag OVA, animals received an i.p. injection of 20 mg OVA
(Sigma-Aldrich, Seelze, Germany), suspended in 2.25 mg aluminum hydroxide (Imject Alum; Pierce, Rockford, IL), in a total volume of 100 ml
on days 0 and 14.
To analyze the effects of lung-specific overexpression of Wnt (Fig. 1A-1),
Wnt/CCSP animals received doxycycline (DOX; 2 mg/ml sterile water;
Sigma-Aldrich) (24) in drinking water starting from day 21 until the end of
the experiment. Allergic airway disease was induced by challenge via the
airways on days 28–30, whereby an OVA solution (1% in PBS) was
nebulized daily for 20 min with an ultrasonic nebulizer [NE-U17; Omron,
Hoofdorp, The Netherlands]. Readouts were performed 48 h following the
last challenge. To analyze the effect of Wnt upregulation in animals with
already established airway disease, a secondary challenge protocol was
used (Fig. 1A-2) (25). In this protocol, animals were not exposed to the
allergen for 6 wk following the first challenge phase; subsequently, they
received a single secondary challenge via the airways by nebulization. To
analyze the function of Wnt, CCSP-rtTA 3 tetO-Wnt1 animals received
DOX treatment starting 7 d before the secondary challenge until the end of
the experiment. Analysis of the animals was performed 48 h following the
secondary challenge.
To assess whether upregulation of the Wnt metabolite b-catenin has
a protective effect in wild-type animals, C57BL/6 animals were sensitized
and challenged as described in Fig. 1A. Instead of inducing Wnt expression, animals received an i.p. injection of LiCl (dissolved in sterile water;
Sigma-Aldrich; 200 mg/kg body weight/d) daily, starting 10 d before the
first challenge (Fig. 1B).
To analyze the effects of a lung-specific upregulation of Wnt on DC
migration, animals were anesthetized by i.p. injection of ketamine/rompun
(Ketamin-ratiopharm; Ratiopharm, Ulm, Germany/Rompun 2%; Bayer,
Leverkusen, Germany) and received a single intranasal (i.n.) application of
fluorescence-labeled OVA derivate (OVA Alexa Fluor 647; MoBiTec,
Göttingen, Germany) dissolved in 40 ml PBS (26, 27). At 12, 24, and 48 h
following treatment, regional lymph nodes were collected and analyzed via
FACS for OVA+ cells. Wnt overexpression was induced by DOX feeding
of CCSP-rtTA 3 tetO-Wnt1 animals again starting 7 d before the i.n.
challenge (Fig. 1C-1).
To further identify the role of DCs in Wnt-mediated events, sensitized
animals received in vitro–generated and OVA-pulsed bone marrow–derived DCs (BMDCs) i.n. on day 28 (Fig. 1C-2); animals were analyzed on
day 30.
Each experiment was performed at least twice, and at least three to five
animals were used per group.
FIGURE 1. Treatment protocols. (A-1) Acute/prophylactic model: days
0 and 14 i.p. injection of OVA/alum; days 28–30 challenge by nebulization
with 1% OVA solution; 7 d prior to challenge start daily DOX application
by feeding; day 32 assay. (A-2) Secondary challenge/therapeutic model:
days 0 and 14 i.p. injection of OVA/alum; days 28–30 challenge by nebulization with 1% OVA solution; 6-wk resting phase; secondary challenge
by single nebulization with OVA; 7 d prior to secondary challenge start
DOX feeding. (B) Lithium model: days 0 and 14 i.p. injection of OVA/
alum; days 28–30 challenge by nebulization with 1% OVA solution; 10 d
prior to challenge daily i.p. injection of LiCl. (C-1) DC migration model:
days 0 and 14 i.p. injection of OVA/alum; day 28 i.n. application of
fluorescence-labeled OVA; 7 d prior to challenge daily DOX application by
feeding. (C-2) DC vaccination model: days 0 and 14 i.p. injection of OVA/
alum; day 28 i.n. application of OVA-loaded DCs; 7 d prior to DC instillation daily DOX application by feeding.
The Journal of Immunology
at least four investigators. Scores ranged from 0 to 5, where 0 was no
detectable brown staining and 5 was highly positive.
FACS analysis
FIGURE 2. Lung-specific induction of Wnt-1 signaling during challenge phase reduced the induction of
an acute model of allergic airway
disease. (A) Airway resistance and
compliance in challenged only animals (unsens), sensitized and challenged WT animals (sens), and
sensitized and challenged DOXtreated CCSP-rtTA 3 tetO-Wnt1
mice (sens + DOX). (B) Cellular
composition of BALF in challenged
only animals treated with DOX
(unsens + DOX), sensitized and
challenged (sens), and sensitized and
challenged animals treated with DOX
(sens + DOX). (C) H&E-stained lung
slices and inflammatory score for
unsens, sens, and sens + DOX groups
(original magnification 3100). (D)
PAS-stained lung slides and analysis of
number of PAS+ cells per millimeter of
basal membrane in unsens, sens, and
sens + DOX animals (original magnification 3400). (E) Titers of OVAspecific IgE, IgG1, and IgG2b in sera
of unsens (sens 2 chall +), sens (sens +
chall + DOX 2), and sens + DOX
(sens + chall + DOX +) mice by
ELISA. All data are mean 6 SEM (n =
8–9 mice/group from two independent
experiments). *p # 0.05, **p # 0.01,
ANOVA. ns, not significant.
and CD103+ DCs, doublets were excluded, and CD11c+ MHCII+ cells were
gated. Within these, OVA+ cells were analyzed further. Following exclusion
of Ly6c+ cells, the proportions of CD103+ and CD11b+ cells were determined. The total number of OVA+ CD11c+ MHCII+ B2202 CD103+ and
OVA+ CD11c+ MHCII+ CD11b+ DCs was calculated by multiplying the
fraction of positive cells by the total cell number (Supplemental Fig. 1). To
determine the amount of plasmacytoid DCs (pDCs), OVA+ cells were analyzed following the exclusion of doublets. pDCs were characterized as
B220+ CD11c+ Gr-1+ cells (30). Again, the total number of OVA+ CD11c+
MHCII+ B220+ CD11c+ Gr-1+ DCs was calculated by multiplying the
fraction of positive cells by the total cell number.
To analyze Tregs, adjusted single-cell suspensions of either lung or tLNs
were stained with PerCP-Cy5.5–labeled anti-mouse CD3, PeCy7–conjugated anti-mouse CD4, and PE-labeled anti-mouse CD25 (all from BD
Biosciences). Following incubation and washing, intracellular staining
against Foxp3 was performed by using the Foxp3 Staining Buffer Set
(eBioscience). After fixation and permeabilization, Foxp3 was stained by
incubation with allophycocyanin-labeled anti-mouse Foxp3 Ab.
FACS measurements were performed on a FACSCanto II (BD Biosciences) using Diva software. Final analysis of FACS data and graphics was
performed using FlowJo software (TreeStar, Ashland, OR).
Ag stimulation of lung and lymph node single cells in vitro
To assess in vitro Ag-specific cytokine production of lung and lymph node
cells, single-cell suspensions of lung or lymph nodes were prepared under
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
FACS analysis was used to assess the migratory behavior of conventional DC,
T cells, and regulatory T cell (Treg) populations in lung and draining lymph
nodes (tLNs) ex vivo, as well as DC activation and T cell proliferation in vitro.
To analyze OVA+ DCs in the tLNs, single-cell preparations of tLNs and
stained as described elsewhere (26). In short, tLNs were disrupted and
transferred over a cell strainer into a 5-ml round-bottom tube (Becton
Dickinson, Heidelberg, Germany). Cells were washed, and cell count was
determined. Cell numbers were adjusted to 2 3 107 cells/ml FACS washing
buffer, and 1 3 106 cells were used for each staining. Unspecific binding was
blocked with FcR-blocking Abs (anti-CD16/CD32; BD Biosciences, Heidelberg, Germany). To identify DC populations, the cells were stained with
FITC-labeled anti–MHC class II (eBioscience, San Diego, CA) and PElabeled anti-mouse CD11c (BD Biosciences). To exclude B cells and further subdivide DC populations, cells were additionally stained with PerCPCy5.5–labeled anti-mouse B220 (BD Biosciences). Among the B2202 cells,
the CD11c/MHCII+ proportion of OVA Alexa Fluor 647+ cells was determined. To further analyze DC subpopulations, additional staining was performed using PE-labeled anti-mouse CD103, PerCP-Cy5.5–labeled antimouse CD11b, PeCy7-labeled anti-mouse CD11c, V450-labeled antimouse Ly6c, PE-labeled anti-mouse F4/80 (all from BD Biosciences) and
Brilliant Violet–labeled anti-mouse Gr-1 (BioLegend). To analyze CD11b+
3
4
sterile conditions. Lungs were cut and transferred into a 50-ml vial. Collagenase type I (0.5 mg/ml; Sigma-Aldrich) was added and, following an
incubation time of 45 min at 37˚C in a shaking water bath, cells were
resuspended at least three times through a cannula in a 10-ml syringe.
Cells were transferred over a cell strainer (70 mm; BD Biosciences) into
a new vial and were treated with Gey’s solution to eliminate RBCs.
Subsequently, cells were washed twice, and cell count was determined.
Lymph node single-cell preparations were made as described elsewhere in
this section. Lung and lymph node single-cell preparations were adjusted
to a concentration of 1 3 107 cells/ml test medium (TM; IMDM + 10%
FCS [both from PAA], 1% Pen-Strep [Sigma-Aldrich]). Cells were incubated or not for 72 h with 250 mg/ml OVA (Sigma-Aldrich; Grade V) at
37˚C. Following incubation, supernatants were harvested and stored at 220˚C
for further analysis.
Ag-specific and cytokine ELISA
Adoptive transfer of Ag-pulsed BMDCs
In short, following lysis and washing, bone marrow cells were incubated in
culture medium (IMDM, 10% FCS, 50 mM 2-ME, 2 mM glutamine, 100
mg/ml streptomycin, 100 U/ml penicillin supplemented with 5% GM-CSF)
for 8 d to obtain BMDCs. On day 8, BMDCs were incubated with 100 mg/ml
FIGURE 3. Induction of Wnt-1 during re-exposure of Ag reduces the inflammatory response
in a secondary challenge model. (A) Airway resistance in sensitized and challenged/rechallenged animals (sens), sensitized and challenged/
rechallenged animals with induced Wnt-1 expression (sens + DOX), and unsensitized challenged/rechallenged animals (unsens + DOX).
(B) BALF composition of unsensitized challenged/rechallenged animals (unsens + DOX),
sensitized and challenged/rechallenged animals
(sens), and sensitized and challenged/rechallenged animals with Wnt-1 induction (sens +
DOX). Histological inflammation score (C),
PAS+ cells per millimeter of basal membrane
(D), and titer of OVA-specific Igs IgE, IgG1, and
IgG2b (E) in the sera of unsensitized challenged/
rechallenged (sens 2), sensitized and challenged/rechallenged (sens + DOX 2), and sensitized and challenged/rechallenged treated with
induced Wnt-1 expression (sens + DOX +) animals. Data are mean 6 SEM (n = 8–11 mice/
group from two independent experiments). *p #
0.05, **p # 0.01, ***p # 0.001, ANOVA. ns,
not significant.
OVA overnight. As described in Fig. 1, 1 3 106 cells diluted in 80 ml
PBS was administered i.n. in sensitized animals that were treated or not
with DOX. Readouts were performed 48 h following DC administration.
Unsensitized animals were used as a negative control.
In vitro assays
BMDCs were generated as described (32). On day 7, immature BMDCs
were treated with OVA (5 mg/ml; Merck-Calbiochem; Darmstadt, Germany), and cells were incubated overnight. Subsequently, cells were exposed to various concentrations of recombinant human Wnt-1 (1, 10, 100
ng/ml; BioVision, Milpitas, CA), the GSK-3b–blocking agents LiCl (5, 10,
15 mM) or SB216763 (SB; 1, 5, 10 mM; Enzo Life Sciences, Lörrach,
Germany), or medium alone (control) for 24 h. To compare the effects of
activated versus immature BMDCs, a part of each group was incubated
with LPS for 60 min following treatment with the ligands. The TLR4 ligand LPS (1 mg/ml; Calbiochem/Merck, Darmstadt, Germany) was added
to the cultures of the appropriate groups. After 24 h, DCs were stained for
the expression of CD11c and MHC class II, as described, or stained with
PE-conjugated anti-mouse CD80 (BD Biosciences) and anti-mouse CD86
(eBioscience), as well as with allophycocyanin-conjugated anti-mouse
CD40 and anti-mouse CCR7 (both from BD Biosciences). Expression
patterns were assessed by FACS and analyzed via FlowJo software.
To analyze effects on T cell proliferation, OVA-loaded BMDCs were
generated as described before and treated with ligands. Furthermore, OVABMDC cultures were split in half and activated by adding LPS or by incubating in medium alone. Twenty-four hours following treatment, DCs
were cocultured for 72 h with CD4+ CFSE-labeled OVA-transgenic T cells.
Splenocytes collected from spleens of DO10.11 animals were purified
using MACS Separator–LS columns (Miltenyi Biotec, Bergisch Gladbach,
Germany), biotinylated anti-mouse CD4 (clone H129.19; Institute for
Immunology, Mainz, Germany), and streptavidin-coated beads to obtain
a CD4+ cell population. Purifications were performed according to the
manufacturer’s instructions. Cells were adjusted to 1 3 107 cells/ml, and
carboxyfluorescein diacetate succinimidyl ester (Invitrogen Life Technol-
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
Serum was obtained 48 h after the last challenge. OVA-specific IgG1 and
IgG2b titers were determined by ELISA (BD Pharmingen), following the
manufacturer’s protocol, as described (28). All Abs were used in concentrations recommended by the manufacturer. OVA-specific IgE titers
were analyzed following the protocol, as described (31). The Ab titer was
defined as the reciprocal serum dilution yielding an absorbance reading of
OD = 0.2 after linear-regression analysis.
To determine cytokine concentrations in BALF and supernatants of
restimulated lymph node and lung cells, cytokine levels were analyzed
according to the manufacturer’s instructions for IL-4, IL-5, IFN-g, and IL10 (all from BD Pharmingen), and IL-13 (R&D Systems).
Wnt/b-CATENIN ATTENUATES ASTHMA
The Journal of Immunology
ogies, Darmstadt, Germany) was added to a final concentration of 2.5 mM.
Cells were incubated for 10 min at 37˚C. The reaction was stopped by
addition of 10% FCS. Following centrifugation, cells were washed twice
with 2% FCS and adjusted to 5 3 105 cells/ml in TM. Likewise, BMDCs
were adjusted to 1 3 105/ml, and equal volumes of the cells were plated
out together. Cocultures were established in TM, with and without secondary application of Wnt-1, LiCl, or SB in the same doses as before.
After 72 h, cells were transferred to 1.5-ml reaction tubes and centrifuged.
Supernatants were collected in new tubes for IFN-g measurements via
ELISA. Cells were resuspended in FACS buffer, split, and surface stained
with PerCP-Cy5.5–labeled anti-mouse CD3, Pe-Cy7–conjugated antimouse CD4, PE-conjugated anti-mouse CD25, and allophycocyaninconjugated CD44 (all from BD Biosciences). FACS measurements were
performed on FACSCanto II (BD Biosciences) using Diva software. Final
analysis of FACS data and graphics was achieved using FlowJo software.
Percentage inhibition of cytokine production/proliferation was calculated as
described (33), in short: 100 2 ([comparison example/positive control] 3 100).
For the calculation of proliferation, samples were adjusted to the d mean
fluorescent intensity of untreated and positive control cells.
Statistical analysis
Results
Expression of Wnt suppresses the development of allergic
airway disease
To analyze the role of Wnt during the development of allergic airway
disease, a model of lung-specific DOX-inducible overexpression of
FIGURE 4. Blockade of b-catenin
degradation by application of LiCl
during challenge prevents the development of allergic airway disease in
C57BL/6 mice. To assess the effects
of b-catenin degradation blockade in
wild-type C57BL/6 mice, they were
sensitized and challenged with OVA.
Parameters of allergic airway disease
were compared among animals sensitized and challenged (sens), those additionally treated with LiCl before
challenge (sens + LiCl), and appropriate
controls (unsens/unsens + LiCl). Treatment of sensitized and challenged animals with LiCl reduced all features
of the asthmatic phenotype. (A) AHR
of sens animals, sens + LiCl animals,
unsens animals, and unsens + LiCl animals. (B) Composition of BALF. Inflammatory score (C), mucus-producing
PAS+ cells per millimeter of basal membrane (D), and titers of OVA-specific
IgE, IgG1, and IgG2b (E) in unsensitized
(sens 2), sensitized but not challenged
(sens + chall 2), sensitized and challenged (sens + chall +) and sensitized
and challenged treated with LiCl (sens +
chall + LiCl +) animals. Data are mean 6
SEM (n = 15 mice/group from three
independent experiments). **p # 0.01,
***p # 0.001, ANOVA. ns, not significant.
Wnt was used (Fig. 1A-1). Wnt-1 signaling prevents the cytoplasmic degradation of b-catenin. To analyze whether DOX application
leads to efficient accumulation of b-catenin in the lungs of CCSPrtTA 3 tetO-Wnt1 animals, lung slices were analyzed for b-catenin
expression. Indeed, CCSP-rtTA 3 tetO-Wnt1 mice treated with
DOX showed an increased expression of b-catenin mainly in the
airway epithelium in comparison with mice that were sensitized but
not treated with DOX (Supplemental Fig. 1).
Systemic sensitization of CCSP-rtTA 3 tetO-Wnt1 mice with
the model allergen OVA and subsequent aerosol exposure resulted
in several characteristic features of allergic airway disease, including airway and lung inflammation, airway hyperresponsiveness
(AHR), mucus cell metaplasia, and increased levels of OVAspecific IgE and IgG1 (Fig. 2). Interestingly, treatment of sensitized CCSP-rtTA 3 tetO-Wnt1 mice with DOX, starting 7 d prior
to the first challenge (sens + DOX), resulted in an attenuation of
AHR, eosinophilia in the BALF, and a reduction in mucus cell
numbers (Fig. 2). Lung tissue inflammation was reduced slightly in
CCSP-rtTA 3 tetO-Wnt1 mice with DOX; however the difference
compared with CCSP-rtTA 3 tetO-Wnt1 mice without DOX did
not reach statistical significance. To assess whether DOX treatment
has any effect on the allergic airway disease, sensitized and challenged wild-type animals treated or not with DOX were analyzed.
No direct effect of DOX on the development of allergic airway
disease was observed. Sensitized wild-type animals treated with
DOX showed no significant differences in the degree of airway
inflammation or AHR following allergen exposure compared with
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
ANOVA was used to determine the levels of difference among all groups.
Comparisons for all pairs were performed using the unpaired Student t test.
The level of significance for p values was set at 0.05. Values for all
measurements are expressed as mean 6 SEM.
5
6
sensitized and challenged wild-type controls left untreated
(Supplemental Fig. 2). Wnt-1 overexpression in the lung and the
associated reduction in allergen-induced inflammation did not
affect systemic sensitization, as evidenced by unchanged OVAspecific IgE, IgG1, and IgG2b levels in sera, irrespective of DOX
treatment (Fig. 2E).
Increased Wnt-1 expression decreases airway inflammation in
a secondary allergen challenge model
Treatment of wild-type mice with LiCl protects them from
development of allergic airway disease
LiCl is known to prevent b-catenin degradation and, therefore,
mimics canonical Wnt signaling (34). To analyze whether the Wnt1–induced regulatory effects in CCSP-rtTA 3 tetO-Wnt1 mice are
achievable in wild-type animals, C57BL/6 animals were treated
with LiCl (Fig. 1B). Sensitized mice were exposed to LiCl 10 d
prior to the challenge, as described in Materials and Methods. AHR
and airway inflammation were assessed 48 h following the last allergen exposure (Fig. 1C). Application of LiCl resulted in a reduced
AHR in comparison with untreated mice (Fig. 4A). Furthermore,
numbers of eosinophils in the BALF, influx of inflammatory cells in
the lung tissue, and goblet cell metaplasia also were significantly
decreased in the LiCl-treated animals (Fig. 4B–D). However,
treatment with LiCl had basically no effect on sensitization because
serum levels of OVA-specific Igs remained unaffected (Fig. 4E). Yet,
LiCl treatment reduced concentrations of Th2 cytokines, such as IL-4
and IL-5, in BALF, as well as in supernatants of unstimulated and
restimulated cells isolated from regional lymph nodes compared with
levels of the respective control group (Supplemental Fig. 3A, 3B).
Treatment with LiCl had no effect on the frequency of Tregs (defined
as CD4+ CD25+ Foxp3+ cells) (Supplemental Fig. 3C, 3D).
Wnt overexpression does not affect induction of Tregs but does
affect the migratory behavior of Ag-loaded DCs
Stabilization of b-catenin induced by Wnt-1 has been associated with
increased survival of Tregs (35). Therefore, the number of Tregs
(defined as CD4+ CD25+ Foxp3+ cells) was assessed in lungs and
tLNs of animals following secondary airway challenge. However,
there were no significant differences in the frequencies and absolute
numbers of Tregs in either the lungs or lymph nodes of sensitized and
challenged CCSP-rtTA 3 tetO-Wnt1 mice that did or did not receive
DOX (Fig. 5, data not shown). This suggests that significant expansion of Tregs does not occur following Wnt overexpression.
To further assess the mechanisms underlying the suppressive
effect of Wnt on the development of allergic airway disease, DC
migration was investigated following allergen challenge. To this
FIGURE 5. Frequencies of Tregs are unaffected in mice overexpressing Wnt-1 in lung. Lungs and tLNs were analyzed for Tregs (characterized as CD4+
CD25+ Foxp3+ cells by FACS). Percentage of CD25+ Foxp3+ Tregs within CD4+ T cell population in tLNs (A) or lungs (B). Dot plots show Foxp3 versus
CD25 staining among CD4+ cells. Data are mean 6 SEM (n = 4–6 mice/group for tLN analysis and n = 6 mice/group for lung analysis from two independent experiments). ns, not significant.
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
Overexpression of Wnt-1 reduced allergic airway disease in a primary, prophylactic model of allergic airway disease. The effect of
Wnt-1 overexpression also was assessed in a more therapeutic model
of secondary allergen challenge (Fig. 1A-2). To this end, CCSP-rtTA 3
tetO-Wnt1 mice were sensitized and then challenged with nebulized
OVA to induce allergic airway disease. Five weeks later, CCSPrtTA 3 tetO-Wnt1 mice received either DOX to induce Wnt-1
expression or PBS as a control. After another week, one secondary challenge was performed. As expected, secondary airway
challenge induced AHR, airway inflammation, and mucus cell
metaplasia in CCSP-rtTA 3 tetO-Wnt1 mice that received PBS.
Similar to the results in the primary airway challenge model, application of DOX reduced AHR, airway inflammation, and mucus
cell numbers (Fig. 3A–D). Again, systemic levels of OVA-specific
Igs were unaffected by the administration of DOX, because OVAspecific IgE, IgG1, and IgG2b titers were comparable in all sensitized animals (Fig. 3E).
Wnt/b-CATENIN ATTENUATES ASTHMA
The Journal of Immunology
end, sensitized CCSP-rtTA 3 tetO-Wnt1 mice were exposed to
a single i.n. application of the fluorescent OVA derivative OVA–
Alexa Fluor 647 (Fig. 1C-1). At 12, 24, and 48 h following application, tLNs were isolated and analyzed by FACS for immigration
of OVA+ B2202 CD11c+ MHCII+ DCs (Fig. 6A, Supplemental
Fig. 1). Similar to previous results (26), the numbers of fluorescent DCs peaked at 24 h after the application of labeled Ag (Fig.
6B, 6C). Interestingly, pretreatment of mice with DOX resulted in
a significant reduction in fluorescent cells in regional lymph nodes,
comparable to the numbers found in nonsensitized animals. Further
characterization of the migratory DCs showed that MHCII+ CD11c+
CD11b+ DCs and MHCII+ CD11c+ CD103+ DCs were decreased in
tLNs in DOX-exposed animals following allergen exposure compared with sensitized and challenged animals that did not receive
DOX (Fig. 6D, Supplemental Fig. 1). However, this decrease was
only statistically significant for MHCII+ CD11c+ CD11b+ DCs. The
numbers of MHCII+ CD11c+ B220+ GR1+ DCs were low and were
not statistically different among all experimental groups (Fig. 6D).
To further analyze whether the regulatory effect of Wnt is mediated
by DCs, ex vivo–generated wild-type DCs were loaded with Ag
and applied prior to airway challenge to DOX-exposed animals.
FIGURE 6. Wnt-1 expression influences the migration of Ag-loaded DCs. To assess DC migration,
animals were challenged by i.n. application of
OVA–Alexa Fluor 647 (OVA-Alexa647). DCs were
characterized as CD11c+ MHCII+ B2202 cells by
FACS. Among this population, the number of OVAAlexa647+ cells was determined. (A) FACS plots
show CD11c+ and MHCII+ cells among the population of B2202 cells in the tLN at 24 h following
i.n. treatment (upper panels). Graphs depict OVAAlexa647+ cells within the CD11c+ MHCII+ DC
population (lower panels). White areas represent
the respective CCSP*Wnt-1 unsens + DOX,
CCSP*Wnt-1 sens 2 DOX, and CCSP*Wnt-1 sens +
DOX groups, whereas the light grey areas denote
the OVA-Alexa6472 control group. (B) Migration
kinetics of OVA-Alexa647+ DCs were analyzed by
calculating the total numbers of OVA-Alexa647+
DCs at 12, 24, and 48 h following i.n. application of
OVA-Alexa647. Sensitized and challenged animals
without DOX treatment are denoted by filled circles; animals also treated with DOX are denoted by
open circles. (C) Total number of OVA-Alexa647+
DCs in the tLNs 24 h following application of
Alexa Fluor 647. For (B) and (C), data are mean 6
SEM (n = 8 mice/group from two independent
experiments for the 24-h time point, and n = 4
animals/group from one experiment for the 12- and
48-h time points). (D) Total number of OVA+ DC
subpopulations in the tLN. The total number of
OVA+ CD11c+ MHCII+ Ly6c+ CD11b+ DCs, OVA+
CD11c+ MHCII+ Ly6c+ CD103+ DCs, and OVA+
B220+ CD11c+ Gr-1+ pDCs. Data represent cell
numbers for unsensitized WT (white bars), sensitized WT (diagonal striped bar), and unsensitized
(black bar) and sensitized (horizontal striped bar)
CCSP-rtTA 3 tetO-Wnt1 mice that received DOX.
Mean 6 SEM are given. *p # 0.05, ANOVA.
Following systemic sensitization, CCSP-rtTA 3 tetO-Wnt1 animals received DOX daily to induce Wnt expression, starting 7 d
prior to the application of Ag-pulsed DCs (Fig. 1C-2). Indeed,
a single i.n. application of Ag-pulsed DCs caused increased
numbers of eosinophils in sensitized animals but not in nonsensitized controls (Fig. 7). Interestingly, no significant differences in eosinophil numbers could be detected in sensitized mice
that received DOX compared with mice that did not receive DOX
(Fig. 7A). Similarly, there also was no difference in tissue inflammation or cytokine production in supernatants of unstimulated
cells or stimulated cells from lung and lymph nodes following
application of Ag-pulsed DCs in sensitized animals treated or not
with DOX (Fig. 7B–D). These data suggest that allergen-pulsed
DCs can override the suppressive effect of Wnt-1 in this model.
In vitro effects of Wnt-1/b-catenin signaling on DC/T cell
interaction
In vitro assays were performed to analyze the Wnt-1–mediated effects
on DCs and their interplay with T cells more closely. Unstimulated
and LPS-activated OVA-loaded BMDCs were cocultured with CFSElabeled OVA peptide–specific CD4+ T cells. To assess the effects of
Wnt-1, this factor was added in various concentrations during DC
activation (only DCs) or during the complete DC/T cell coculture
period (total). Addition of Wnt did not affect the expression of co-
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
Transfer of OVA-loaded DCs abrogates Wnt-induced
regulatory effects
7
8
Wnt/b-CATENIN ATTENUATES ASTHMA
stimulatory molecules on DCs (Supplemental Fig. 4A). Interestingly,
OVA-specific T cells incubated with OVA-loaded DCs proliferated
strongly, whereas T cells that were incubated with Wnt-exposed DCs
proliferated significantly less. This was true when Wnt was added
only to the DC culture or to the DC/T cell coculture (Fig. 8A). In
addition, expression of activation markers on T cells and cytokine
production were much lower in T cells that were exposed to Wnttreated DCs compared with untreated DCs (Fig. 8B, 8C).
In agreement with these data, a protective effect of b-catenin accumulation following blockade of GSK-3b also was detected. Activated DCs incubated with LiCl or the highly specific GSK-3b
inhibitor SB prevented T cell proliferation and IFN-g secretion
(Supplemental Fig. 4B, 4C).
Discussion
The present study demonstrated that increased expression of
Wnt-1 in the lung abrogates the development of allergic airway
disease. Induced Wnt-1 overexpression in allergen-sensitized animals before inhaled airway allergen challenge resulted in reduced
numbers of eosinophils in the BALF, AHR, and mucus production.
This effect was found in a more prophylactic primary challenge
model, as well as in a chronic model in which an inflammatory lung
disease already had been established. Similarly, in the latter model,
Wnt-1 overexpression before and during secondary challenge reduced the development of AHR, eosinophilia in the BALF, and
tissue inflammation, confirming the suppressive effect.
Wnt molecules have been ascribed important functions during
lung development and different lung diseases. Wnt/b-catenin
signaling is involved in tissue repair mechanisms in the lung,
because reduced Wnt expression can contribute to increased development of emphysema (20). Additionally, overexpression of
Wnt molecules induces proliferation and migration of fibroblasts
and is involved in lung fibrosis (36, 37). Consequently, inhibition
of the Wnt pathway represents a therapeutic option in bleomycin-
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
FIGURE 7. Ex vivo Ag-loaded DCs overcome in vivo effects of Wnt-1 in the DC vaccination model. Unsensitized and sensitized WT and CCSP-rtTA 3
tetO-Wnt1 (Wnt/CCSP) mice fed DOX to induce Wnt-1 expression were treated i.n. with OVA-loaded BMDCs. The developing inflammatory response was
analyzed. (A) Cellular composition of BALF from unsensitized animals + OVA DCs (WT [white bars] and CSP/Wnt [black bars]) and sensitized animals +
OVA DCs (WT [diagonal striped bars] and CCSP/Wnt [horizontal striped bars]). (B) Inflammation score. Concentrations of cytokines (IL-5, IFN-g, IL-10,
and IL-13) were analyzed in OVA-restimulated single-cell suspensions (gray bars) or in nonstimulated single-cell suspensions (white bars) of the lung (C) or
tLNs (D). Data are mean 6 SEM (n = 4–6 mice/group from one experiment). ns, not significant.
The Journal of Immunology
9
induced fibrosis (38). However, the role of the canonical Wnt
pathway in the development and regulation of allergic airway
disease is not well understood. Gene expression studies in patients
with asthma revealed a correlation between Wnt expression and
the Th2-specific phenotype of this disease. These studies demonstrated that increased mRNA expression of several Wnt cascade
molecules positively correlated with a Th2 signature and impaired
lung function in asthma patients (39, 40). Irrespective of these
gene expression studies, the functional role of Wnt in allergic
airway disease remains unexplained. In the current study, increased expression of Wnt-1 had a protective effect on the development of allergic airway disease. Wnt-1 overexpression did
not affect levels of anti-OVA Igs in sera of sensitized animals. This
indicates that the protective effects are not due to a Wnt-1–de-
pendent systemic modulation of the adaptive immune response but
rather to local effects in the lung that abrogate the development of
the disease. A nonspecific effect of DOX treatment on the reduction of airway inflammation was ruled out, because no reduction in allergic airway disease was detected in sensitized and
challenged wild-type animals exposed to DOX. The relevance of
these findings was further elucidated in a rechallenge model. This
more chronic model confirmed the decrease in airway inflammation, AHR, and mucus cell numbers and demonstrated again that
increased Wnt-1 expression suppressed allergic airway disease,
even in a therapeutic model.
To further support our findings of b-catenin–dependent suppression of allergic disease, pharmacological agents known to
activate b-catenin were applied. Application of the GSK-3b in-
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
FIGURE 8. Wnt-1 affects DC functions in vitro. Immature and mature BMDCs were incubated with OVA; aliquots were treated with different doses of
Wnt-1 and subsequently incubated with OVA peptide–specific T cells. T cell activation was analyzed. (A) OVA-loaded immature or mature DCs were
cocultured with CFSE-labeled CD4+ T cells expressing an OVA peptide specific–transgenic TCR. After 72 h, proliferation of T cells was analyzed by
measuring CFSE fluorescence reduction via FACS. Dot plots depict CD4+ versus CFSE (left panels). Bar graph depicts CFSE fluorescence in the CD4+ T
cell population (right panel). Shaded graph represents T cells cocultured with untreated OVA-loaded immature DCs. The green lines depict T cells coculture with LPS-stimulated OVA-loaded mature DCs. The red lines represent proliferation of Wnt-1–treated T cells. Wnt-1 was supplemented during the
entire incubation period. Bar graph shows the percentage inhibition of T cell proliferation from T cell cocultures with differentially Wnt-1–pretreated
BMDCs in comparison with the positive control (OVA-loaded mature DC + T cells). Wnt-1 was applied only during BMDC culture (only DC) or during
DC/T cell coculture (total). (B) Mean fluorescence intensity (MFI) of CD44 (left panel) or CD25 (right panel) on the surface of CD4+ CFSE+ T cells
cocultured with either BMDC population and pretreated as indicated. (C) Cytokines were detected in the supernatants of the BMDC/T cell cocultures after
72 h of incubation. Bar graph represents the percentage inhibition of IFN-g secretion in the described groups. Two experiments were performed; graphs and
dot plots depict results from one of two independent experiments (n = 2). *p # 0.05, **p # 0.01, ***p # 0.001 versus positive control, t test.
10
liferation, surface expression of activation markers, and cytokine
secretion were all impaired. This suppressive effect of Wnt-1 on
DCs seems to be comparable to the effect of other Wnt molecules.
Wnt-5a suppressed the production of IL-12 and IFN-g in human
PBMCs exposed to microbial compounds (45). Similar to Wnt-1,
inhibition of GSK-3b by LiCl or the highly specific inhibitor SB
resulted in modulation of DC/T cell interactions. SB treatment
caused a suppression of T cell proliferation, but this was only
apparent when the inhibitor was constantly present during the
coculture. Blockade of b-catenin degradation during the DC activation period alone was insufficient to suppress DC-mediated
T cell proliferation. Nevertheless, induction of tolerance is not
necessarily accompanied by a reduction in T cell proliferation.
Also, Ag delivery from DCs under steady-state conditions induced
T cell proliferation, without generation of T cell subtypes and
prolonged activation (46). Moreover, OVA-pulsed immature DCs
induced T cell proliferation, but only mature OVA-pulsed DCs
were able to initiate the production of IFN-g (47). In the current
study, treatment of BMDCs with LiCl or SB was able to prevent
cytokine production of T cells. This phenomenon was observed,
irrespective of whether LiCl or SB was present during the coculture of DCs and T cells or whether DCs were treated before the
onset of the cocultures. These results suggest that Wnt-1, via
GSK-3b blockade, mediated accumulation of b-catenin, which, in
turn, suppressed BMDC activation.
In summary, the present experiments demonstrate in an acute, as
well as in a therapeutic, model that activation of the canonical Wnt1/b-catenin pathway ameliorates the development of allergic airway disease. This effect seems to be linked to the suppression of
DC activation, because overexpression of Wnt-1 decreased migration of DCs to the tLNs and the induction of appropriate T cell
responses. The present data demonstrate that the Wnt-1/b-catenin
pathway represents a novel interesting target to modulate allergic
responses.
Acknowledgments
We thank Julia Altmeier and the FACS Core Facility of the Institute of
Toxicology (University Medical Center Mainz) for technical support and
Pieter Hiemstra and Merete Long (Leiden University Medical Center) for
critical reading of the manuscript.
Disclosures
The authors have no financial conflicts of interest.
References
1. Wenzel, S. E. 2012. Asthma phenotypes: the evolution from clinical to molecular
approaches. Nat. Med. 18: 716–725.
2. Zhang, D. H., L. Yang, L. Cohn, L. Parkyn, R. Homer, P. Ray, and A. Ray. 1999.
Inhibition of allergic inflammation in a murine model of asthma by expression of
a dominant-negative mutant of GATA-3. Immunity 11: 473–482.
3. Gr€unig, G., M. Warnock, A. E. Wakil, R. Venkayya, F. Brombacher,
D. M. Rennick, D. Sheppard, M. Mohrs, D. D. Donaldson, R. M. Locksley, and
D. B. Corry. 1998. Requirement for IL-13 independently of IL-4 in experimental
asthma. Science 282: 2261–2263.
4. Akbari, O., R. H. DeKruyff, and D. T. Umetsu. 2001. Pulmonary dendritic cells
producing IL-10 mediate tolerance induced by respiratory exposure to antigen.
Nat. Immunol. 2: 725–731.
5. Lambrecht, B. N., B. Salomon, D. Klatzmann, and R. A. Pauwels. 1998. Dendritic
cells are required for the development of chronic eosinophilic airway inflammation
in response to inhaled antigen in sensitized mice. J. Immunol. 160: 4090–4097.
6. Hikasa, H., and S. Y. Sokol. 2013. Wnt signaling in vertebrate axis specification.
Cold Spring Harb. Perspect. Biol. 5: a007955.
7. Clevers, H. 2006. Wnt/beta-catenin signaling in development and disease. Cell
127: 469–480.
8. Katoh, M. 2005. WNT/PCP signaling pathway and human cancer. Oncol. Rep. 14:
1583–1588.
9. Kohn, A. D., and R. T. Moon. 2005. Wnt and calcium signaling: beta-cateninindependent pathways. Cell Calcium 38: 439–446.
10. Korinek, V., N. Barker, K. Willert, M. Molenaar, J. Roose, G. Wagenaar,
M. Markman, W. Lamers, O. Destree, and H. Clevers. 1998. Two members of the
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
hibitor LiCl to mimic the effect of canonical Wnt activation (34)
caused similar effects as did DOX-induced Wnt-1 overexpression
in transgenic mice. Comparable with forced overexpression of
Wnt-1, application of LiCl during challenge also led to reduced
development of allergic airway disease in sensitized animals.
Development of AHR, eosinophilia in BALF, airway inflammation, and goblet cell metaplasia were abrogated in animals treated
with LiCl. The conclusions from the present results are in line
with findings in other models of lung disease. The GSK-3b–inhibitory effect of LiCl was investigated in a mouse model of
emphysema; it caused attenuation of the disease (20).
Immunological inflammatory responses are controlled by the
induction of Foxp3+/CD25+ Tregs. In vitro studies demonstrated
that b-catenin supports Treg survival (35). Surprisingly, neither
Wnt-1 overexpression nor LiCl treatment resulted in increased
frequencies of Tregs in the lungs and these animals showed concentrations of the regulatory cytokine IL-10 in BALF and
restimulated lung and lymph node cell suspensions that were
comparable to sensitized and challenged animals. These findings
suggest that Tregs are not pivotally responsible for the observed
reduction in the allergic inflammatory response. DCs are also
critically involved in shaping immune responses; depending on
their state of activation, they induce or suppress Ag-specific immune responses. DC activation is modulated by exogenous and
endogenous signals, and the composition of their microenvironment determines the outcome of the immune response. It was
shown that DCs are required to induce allergic immune responses
in the lungs (5, 41). Ag uptake, processing, and migration to the
tLNs are essential steps for the induction of an adequate T cell
response. The activation state of DCs determines whether Agspecific tolerance or immunity is initiated. In our study, Wnt-1
overexpression in the lung resulted in reduced migration of Agloaded DCs from the lung to the tLNs in sensitized animals following inhaled allergen exposure. These findings suggest that
Wnt-1 suppressed DC activation following allergen challenge and,
thereby, inhibited the development of the allergic response in the
lung. This assumption is further supported by the finding that
application of in vitro Ag-loaded BMDCs overcame the effects of
Wnt-1 overexpression in the lung. When BMDCs were instilled
i.n. to sensitized and challenged animals, airway inflammation,
AHR, and cytokine production developed, irrespective of Wnt-1
overexpression. These observations support the hypothesis that the
Wnt-1/b-catenin pathway suppresses DC activation in the lung.
Studies in the intestine corroborate these findings. Manicassamy
et al. (42) demonstrated that ablation of b-catenin in DCs led to an
enhanced inflammatory phenotype in a mouse model of inflammatory bowel disease. Furthermore they showed that b-catenin
was important to sustain a tolerance-inducing DC phenotype,
thereby maintaining equilibrium between anergy and immune
response. Moreover, a study demonstrated that disruption of Ecadherin on BMDCs, which also results in activation of the
b-catenin pathway, led to tolerogenic DCs that were able to suppress experimental autoimmune encephalomyelitis (43). However,
an effect of Wnt molecules directly on T cell migration and function
cannot be ruled out. Indeed, Wnt molecules can upregulate matrix
metalloproteinases that facilitate the transmigration of T cells into
the tissue (44).
An in vitro model was established to further analyze the effect of
the Wnt-1/b-catenin pathway on DC activation and allergenspecific T cell cross-talk. Interestingly, application of Wnt-1 to
unstimulated or LPS-activated OVA-loaded BMDCs had no effect
on the expression patterns of the costimulatory molecules CD86,
CD80, and CD40. However, BMDCs treated with Wnt-1 had an
impaired ability to induce a T cell response, because T cell pro-
Wnt/b-CATENIN ATTENUATES ASTHMA
The Journal of Immunology
11.
12.
13.
14.
15.
16.
17.
18.
19.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31. Cruz, E. A., S. Reuter, H. Martin, N. Dehzad, M. F. Muzitano, S. S. Costa,
B. Rossi-Bergmann, R. Buhl, M. Stassen, and C. Taube. 2012. Kalanchoe pinnata inhibits mast cell activation and prevents allergic airway disease. Phytomedicine 19: 115–121.
32. Bros, M., N. Wiechmann, V. Besche, T. Castor, S. Sudowe, S. Grabbe, and
A. B. Reske-Kunz. 2009. A novel plasmid DNA electroporation method allows
transfection of murine DC. J. Immunol. Methods 343: 13–20.
33. Dehzad, N., T. Bopp, S. Reuter, M. Klein, H. Martin, A. Ulges, M. Stassen, H.
Schild, R. Buhl, E. Schmitt, and C. Taube. 2011. Regulatory T cells more effectively suppress Th1-induced airway inflammation compared with Th2. J.
Immunol. 186: 2238–2244.
34. Hedgepeth, C. M., L. J. Conrad, J. Zhang, H. C. Huang, V. M. Lee, and
P. S. Klein. 1997. Activation of the Wnt signaling pathway: a molecular
mechanism for lithium action. Dev. Biol. 185: 82–91.
35. Ding, Y., S. Shen, A. C. Lino, M. A. Curotto de Lafaille, and J. J. Lafaille. 2008.
Beta-catenin stabilization extends regulatory T cell survival and induces anergy
in nonregulatory T cells. Nat. Med. 14: 162–169.
36. Königshoff, M., N. Balsara, E. M. Pfaff, M. Kramer, I. Chrobak, W. Seeger, and
O. Eickelberg. 2008. Functional Wnt signaling is increased in idiopathic pulmonary fibrosis. PLoS ONE 3: e2142.
37. Lam, A. P., A. S. Flozak, S. Russell, J. Wei, M. Jain, G. M. Mutlu,
G. R. Budinger, C. A. Feghali-Bostwick, J. Varga, and C. J. Gottardi. 2011.
Nuclear b-catenin is increased in systemic sclerosis pulmonary fibrosis and
promotes lung fibroblast migration and proliferation. Am. J. Respir. Cell Mol.
Biol. 45: 915–922.
38. Kim, T. H., S. H. Kim, J. Y. Seo, H. Chung, H. J. Kwak, S. K. Lee, H. J. Yoon,
D. H. Shin, S. S. Park, and J. W. Sohn. 2011. Blockade of the Wnt/b-catenin
pathway attenuates bleomycin-induced pulmonary fibrosis. Tohoku J. Exp. Med.
223: 45–54.
39. Sharma, S., K. Tantisira, V. Carey, A. J. Murphy, J. Lasky-Su, J. C. Celedón,
R. Lazarus, B. Klanderman, A. Rogers, M. Soto-Quirós, et al. 2010. A role for
Wnt signaling genes in the pathogenesis of impaired lung function in asthma.
Am. J. Respir. Crit. Care Med. 181: 328–336.
40. Choy, D. F., B. Modrek, A. R. Abbas, S. Kummerfeld, H. F. Clark, L. C. Wu,
G. Fedorowicz, Z. Modrusan, J. V. Fahy, P. G. Woodruff, and J. R. Arron. 2011.
Gene expression patterns of Th2 inflammation and intercellular communication
in asthmatic airways. J. Immunol. 186: 1861–1869.
41. Hammad, H., and B. N. Lambrecht. 2008. Dendritic cells and epithelial cells:
linking innate and adaptive immunity in asthma. Nat. Rev. Immunol. 8: 193–204.
42. Manicassamy, S., B. Reizis, R. Ravindran, H. Nakaya, R. M. Salazar-Gonzalez,
Y. C. Wang, and B. Pulendran. 2010. Activation of beta-catenin in dendritic cells
regulates immunity versus tolerance in the intestine. Science 329: 849–853.
43. Jiang, A., O. Bloom, S. Ono, W. Cui, J. Unternaehrer, S. Jiang, J. A. Whitney,
J. Connolly, J. Banchereau, and I. Mellman. 2007. Disruption of E-cadherinmediated adhesion induces a functionally distinct pathway of dendritic cell
maturation. Immunity 27: 610–624.
44. Wu, B., S. P. Crampton, and C. C. Hughes. 2007. Wnt signaling induces matrix
metalloproteinase expression and regulates T cell transmigration. Immunity 26:
227–239.
45. Blumenthal, A., S. Ehlers, J. Lauber, J. Buer, C. Lange, T. Goldmann, H. Heine,
E. Brandt, and N. Reiling. 2006. The Wingless homolog WNT5A and its receptor Frizzled-5 regulate inflammatory responses of human mononuclear cells
induced by microbial stimulation. Blood 108: 965–973.
46. Hawiger, D., K. Inaba, Y. Dorsett, M. Guo, K. Mahnke, M. Rivera, J. V. Ravetch,
R. M. Steinman, and M. C. Nussenzweig. 2001. Dendritic cells induce peripheral
T cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 194:
769–779.
47. de Heusch, M., G. Oldenhove, J. Urbain, K. Thielemans, C. Maliszewski,
O. Leo, and M. Moser. 2004. Depending on their maturation state, splenic
dendritic cells induce the differentiation of CD4(+) T lymphocytes into memory
and/or effector cells in vivo. Eur. J. Immunol. 34: 1861–1869.
Downloaded from http://www.jimmunol.org/ by guest on January 20, 2017
20.
Tcf family implicated in Wnt/beta-catenin signaling during embryogenesis in the
mouse. Mol. Cell. Biol. 18: 1248–1256.
van Es, J. H., P. Jay, A. Gregorieff, M. E. van Gijn, S. Jonkheer, P. Hatzis,
A. Thiele, M. van den Born, H. Begthel, T. Brabletz, et al. 2005. Wnt signalling
induces maturation of Paneth cells in intestinal crypts. Nat. Cell Biol. 7: 381–386.
Lowry, W. E., C. Blanpain, J. A. Nowak, G. Guasch, L. Lewis, and E. Fuchs.
2005. Defining the impact of beta-catenin/Tcf transactivation on epithelial stem
cells. Genes Dev. 19: 1596–1611.
Reya, T., A. W. Duncan, L. Ailles, J. Domen, D. C. Scherer, K. Willert, L. Hintz,
R. Nusse, and I. L. Weissman. 2003. A role for Wnt signalling in self-renewal of
haematopoietic stem cells. Nature 423: 409–414.
Hartmann, C. 2006. A Wnt canon orchestrating osteoblastogenesis. Trends Cell
Biol. 16: 151–158.
Takada, I., A. P. Kouzmenko, and S. Kato. 2009. Wnt and PPARgamma signaling in osteoblastogenesis and adipogenesis. Nat. Rev. Rheumatol. 5: 442–447.
Königshoff, M., and O. Eickelberg. 2010. WNT signaling in lung disease:
a failure or a regeneration signal? Am. J. Respir. Cell Mol. Biol. 42: 21–31.
Shu, W., Y. Q. Jiang, M. M. Lu, and E. E. Morrisey. 2002. Wnt7b regulates
mesenchymal proliferation and vascular development in the lung. Development
129: 4831–4842.
Li, C., J. Xiao, K. Hormi, Z. Borok, and P. Minoo. 2002. Wnt5a participates in
distal lung morphogenesis. Dev. Biol. 248: 68–81.
Winn, R. A., L. Marek, S. Y. Han, K. Rodriguez, N. Rodriguez, M. Hammond,
M. Van Scoyk, H. Acosta, J. Mirus, N. Barry, et al. 2005. Restoration of Wnt-7a
expression reverses non-small cell lung cancer cellular transformation through
frizzled-9-mediated growth inhibition and promotion of cell differentiation. J.
Biol. Chem. 280: 19625–19634.
Kneidinger, N., A. O. Yildirim, J. Callegari, S. Takenaka, M. M. Stein,
R. Dumitrascu, A. Bohla, K. R. Bracke, R. E. Morty, G. G. Brusselle, et al. 2011.
Activation of the WNT/b-catenin pathway attenuates experimental emphysema.
Am. J. Respir. Crit. Care Med. 183: 723–733.
Königshoff, M., M. Kramer, N. Balsara, J. Wilhelm, O. V. Amarie, A. Jahn,
F. Rose, L. Fink, W. Seeger, L. Schaefer, et al. 2009. WNT1-inducible signaling
protein-1 mediates pulmonary fibrosis in mice and is upregulated in humans with
idiopathic pulmonary fibrosis. J. Clin. Invest. 119: 772–787.
Perl, A. K., J. W. Tichelaar, and J. A. Whitsett. 2002. Conditional gene expression in the respiratory epithelium of the mouse. Transgenic Res. 11: 21–29.
Gunther, E. J., S. E. Moody, G. K. Belka, K. T. Hahn, N. Innocent, K. D. Dugan,
R. D. Cardiff, and L. A. Chodosh. 2003. Impact of p53 loss on reversal and
recurrence of conditional Wnt-induced tumorigenesis. Genes Dev. 17: 488–501.
Schonig, K., and H. Bujard. 2003. Generating conditional mouse mutants via
tetracycline-controlled gene expression. Methods Mol. Biol. 209: 69–104.
Taube, C., C. Duez, Z. H. Cui, K. Takeda, Y. H. Rha, J. W. Park, A. Balhorn,
D. D. Donaldson, A. Dakhama, and E. W. Gelfand. 2002. The role of IL-13 in
established allergic airway disease. J. Immunol. 169: 6482–6489.
Reuter, S., N. Dehzad, H. Martin, A. Heinz, T. Castor, S. Sudowe, A. B. ReskeKunz, M. Stassen, R. Buhl, and C. Taube. 2010. Mast cells induce migration of
dendritic cells in a murine model of acute allergic airway disease. Int. Arch.
Allergy Immunol. 151: 214–222.
Reuter, S., N. Dehzad, H. Martin, L. Böhm, M. Becker, R. Buhl, M. Stassen, and
C. Taube. 2012. TLR3 but not TLR7/8 ligand induces allergic sensitization to
inhaled allergen. J. Immunol. 188: 5123–5131.
Reuter, S., A. Heinz, M. Sieren, R. Wiewrodt, E. W. Gelfand, M. Stassen,
R. Buhl, and C. Taube. 2008. Mast cell-derived tumour necrosis factor is essential for allergic airway disease. Eur. Respir. J. 31: 773–782.
Bopp, T., N. Dehzad, S. Reuter, M. Klein, N. Ullrich, M. Stassen, H. Schild,
R. Buhl, E. Schmitt, and C. Taube. 2009. Inhibition of cAMP degradation
improves regulatory T cell-mediated suppression. J. Immunol. 182: 4017–4024.
Nakano, H., M. Yanagita, and M. D. Gunn. 2001. CD11c(+)B220(+)Gr-1(+)
cells in mouse lymph nodes and spleen display characteristics of plasmacytoid
dendritic cells. J. Exp. Med. 194: 1171–1178.
11