結案報告

advertisement
Induced apoptosis of Th2 lymphocytes and inhibition of
airway hyperresponsiveness and inflammation by
combined lactic acid bacteria treatment
1
2
3
4
5
Wen-Hsin Lina, Chi-Rei Wub, Hong-Zin Leea, Yueh-Hsiung Kuoa, Hung-Shin Wena, Tze-Yi Linc,
6
Chia-Ying Leed, Shi-Ying Huange*, Ching-Yuang Lind,f*
7
8
a
School of Pharmacy, b Institute of Chinese Pharmaceutical Sciences, School of Pharmacy, China
9
Medical University, Taichung, Taiwan
10
c
Department of pathology, China Medical University Hospital, Taichung, Taiwan
11
d
College of Medicine, China Medical University, Taichung, Taiwan
12
e
Department of pediatrics, Armed Force Taoyuan General Hospital, Taoyuan, Taiwan
13
f
Clinical Immunologic Center, Children’s Medical Center, China Medical University Hospital,
14
Taichung, Taiwan
15
16
Running Title:
17
Ameliorate airway hyperresponsiveness by probiotics
18
19
20
* Correspondence and equal correspondence: Ching-Yuang Lin and Shi-Ying Huang.
21
Address for correspondence: Ching-Yuang Lin MD., Ph.D. Clinical Immunogical Center, China
22
Medical University Hospital, 2 Yuh-Der Road, Taichung, 404, Taiwan
23
Tel.: +886-4-22052121#1520; Fax: +886-4-22081079.
24
E-mail address: cylin@mail.cmuh.org.tw
25
1
1
Abstract
2
3
Several lactic acid bacteria (LAB) demonstrably regulate the immune system and inhibit allergic
4
disease. This study examined whether oral feeding of either Lactobacillus paracasei (L. paracasei)
5
BB5 and/or L. rhamnosus BB1 suppress ovalbumin (OVA)-induced airway hyperresponsiveness
6
(AHR) and inflammation in a murine model. OVA-specific immune responses, cell profile of
7
bronchoalveolar lavage fluid (BALF), and airway AHR were assessed following OVA and
8
methacholine challenge. We investigated whether LAB can enhance CD4+FoxP3+ and CD8+FoxP3+
9
regulatory T (Treg) cells in splenic cells and apoptosis of CD4+IL-4+ T cells. Results found oral
10
administration of combined LAB better than single L. paracasei or L. rhamnosus strain, improving
11
Penh ratio after challenge with methacholine. High-dose combined LAB starkly decreased synthesis
12
of OVA-specific IgE and IgG2a levels, as well as eosinophils infiltration in BALF. In addition,
13
CD4+IL-4+ T cells decreased while CD4+FoxP3+ and CD8+FoxP3+ Treg cells increased significantly
14
in splenic mononuclear cells of high-dose combined LAB group. Findings indicate allergen-induced
15
AHR and airway allergic inflammation suppressed by enhances CD4+FoxP3+ and CD8+FoxP3+ Treg
16
populations as well as Th1 cell response after treating with combined LAB. This study may provide a
17
basis for developing a novel therapeutic or protective method for airway allergic disease.
18
19
Key words: lactic acid bacteria, ovalbumin, airway hyperresponsiveness, regulatory T cells, apoptosis
20
2
1
1. Introduction
2
3
Asthma is a Th2-mediated chronic inflammatory disease releasing certain Th2 cytokines like
4
interleukin (IL)-4 and IL-5 while increasing specific immunoglobin (Ig) E or accumulation of mucus
5
and various inflammatory cells [1-4]. Epidemiologic studies indicate positive correlation between
6
allergic diseases and changed microbial exposure patterns [5, 6]. These studies found more complex
7
and intensive bacteria colonized in the intestinal tract of non-allergic versus allergic children [7-9]. In
8
addition, major microbiota of non-allergic children intestine is Lactobacilli and Bifidobacteria genus,
9
but Staphylococcus genus is major in allergic children [9,10].
10
Lactic acid bacteria (LAB) are regard as probiotics beneficial to human and animal. Studies
11
suggest that yogurt containing live LAB potentiates a variety of intestinal immune responses and may
12
yield clinical benefits against bacterial or viral infection [11-14]. Lactobacillus plantarum (L.
13
plantarum) isolated from food is indicated to activate Th1 immune response and prevent influenza A
14
viral infection by virus-specific neutralizing IgA in BALFs and sera [15]. Regular consumption of
15
yogurt containing Lactobacillus bulgaricus (L. bulgaricus) and Streptococcus thermophilus
16
demonstrably improves clinical scores among atopic patients or mice, which portends increased
17
production of interferon -γ (IFN-γ) and decreased level of eosinophils [16, 17]. Also, some LAB
18
strains have been reported as effectively and safely improving quality of life for patients suffering
19
from allergic rhinitis, and shown to inhibit allergen-specific T-cell response [18].
20
Human gastrointestinal tract microbiota has been estimated to consist of at least 400 different
21
species and 1014 microorganisms [19, 20] that are major sources of microbial exposure for humans.
22
These may influence immune homeostasis, depending on the strain. Probiotics are defined as living
23
microorganisms beneficial to health. Evidence reveals strains of Lactobacilli and Bifidobacteria can
24
influence immune function via diverse pathways, including effects on antigen-presenting cells (APC):
25
e.g., circulating monocytes, local dendritic cells (DCs), regulatory T (Treg) cells, effector T and B
3
1
cells or macrophage [21-24]. Studies also find cytokine profiles with obvious differences among
2
strains of LAB co-cultured with human peripheral blood mononuclear cells (PBMC) or other APC
3
[25-28]. Likewise, about 30 potential probiotic strains were isolated for prevention or treatment of
4
immune imbalance disorders [29-31]; some strains of Bifidobacteria also enhanced Th1 responses or
5
induced Treg cells. Other studies found few probiotics stimulating production of INF-γ and inhibiting
6
secretion of allergy-related IL-4, IL-5 and specific-Immunoglobulin E (IgE) production. These
7
indicate potential strains improving symptoms of allergy [32, 33]. Several clinical trials showed
8
probiotics as reducing percentage and symptoms of atopic dermatitis or asthma in babies [34, 35].
9
LAB was shown to promote local immune defenses and induce anti-inflammatory cytokines [23,
10
36-38]. Treatment with screening probiotics demonstrated preventive effects against pathogenic- or
11
chemical-induced colitis by gut barrier function and immunomodulatory efficacy [39-42].
12
Lactobacillus paracasei (L. paracasei) treats human allergic rhinitis, and Bifidobacterium longum (B.
13
longum) is proven to relieve clinical symptoms of Japanese cedar pollinosis [43, 44]. Induction of
14
CD4+CD25+ Treg cells with increased IL-10 and transforming growth factor-β (TGF-β) secretion
15
could maintain immune tolerance to allergen challenge [45-47]. CD4+CD25+ Treg cells could express
16
toll-like receptor (TLR) in response to stimulation and might regulate allergic response [48, 49].
17
Inducible CD8+FoxP3+ Treg cells and TGF-β production were identified recently to maintain
18
homeostasis, promote immune tolerance, and regulate host defense against foreign pathogens or
19
allergens [46, 47, 50]. CD8+FoxP3 Treg cells can suppress cellular proliferation of CD4+ naïve and
20
effector T cell by cell-contact. CD8+CD25+ Treg cells directly induce CD4+CD45ROhigh cell
21
apoptosis [51]. Our study tested whether oral administration of L. paracasei BB5, L. rhamnosus BB1
22
or combination thereof effectively down-regulates serum IgE production, airway inflammation, Th2
23
cytokines and airway hyperresponsiveness (AHR) for sensitized animals. We also tested whether
24
LAB enhances CD4+FoxP3+ and CD8+FoxP3+ Treg population or helps suppress allergen-induced
25
Th2 immune response. This study may elucidate a mechanism for novel immunotherapeutic
4
1
treatment and prevention.
2
3
4
5
2. Methods and materials
6
7
2.1. Mice
8
9
Five-week-old specific pathogen-free BALB/c male mice were purchased from the National
10
Laboratory for Animal Breeding and Research Center at Taipei, then housed in six individual cages
11
at 25°C and 60% relative humidity with a 12-h light cycle, eight mice per group. Basic feed was
12
sterilized Laboratory Rodent Diet 5001 (PMI Nutrition International Inc., Richmond, IN), animal test
13
and protocols approved by the China Medical University Hospital Animal Committee.
14
15
2.2. Bacterial preparation
16
17
LAB of L. paracasei BB5 and L. rhamnosus BB1 were isolated from infant feces and validated
18
with effect for anti-allergy in our laboratory, both strains identified by API 50 CHL kit (Biomerieux,
19
Marcy I’Etoile, France) and Lactobacillus identification system. Meanwhile, strains were confirmed
20
by 16S rRNA gene sequence in the GenBank Database, LAB propagated under anaerobic conditions
21
at 37°C in deMan-Rogosa-Sharpe (MRS) broth (Difco, Detroit, MI) until log phase of growth.
22
Finally, cells of LAB were centrifuged and freeze-dried, viable count of powder adjusted to about
23
1×1010 CFU/g.
24
25
2.3. Study design and ovalbumin-induced asthma
5
1
2
Experimental method was modified from Hong and others [52-54]. Six groups of BALB/c mice
3
were designated to examine effects of LAB and their combination. Naïve control mice were neither
4
sensitized with ovalbumin (OVA) nor given LAB treatment; other groups were challenged with OVA
5
to induce allergic symptoms. To establish a murine model of asthma, these mice were immunized by
6
peritoneal injection with 50 μg OVA (Sigma Chemical, St. Louis, MO) plus 1:1 mixture of
7
incomplete Freund’s adjuvant (Sigma) on Days 14 and 21. Mice were exposed to OVA aerosol
8
inhalation (2% OVA in normal saline solution) for 10 minutes on Days 27-28; hyperresponsiveness
9
of airways was assessed on Day 29 after challenged with different concentration of methacholine
10
(Sigma). During experiment (Days 0-28), mice of each group except naïve controls received orally
11
administration of different samples daily: saline (OVA control group), 3.75 mg L. paracasei BB5
12
(P-1×), 3.75 mg L. rhamnosus BB1 (R-1×), 3.75 mg (P+R,-1×) with 1.875 mg BB5 plus 1.875 mg
13
BB1, and 11.25 mg (P+R,-3×) with three-fold dose of 3.75 mg P+R. Mice were sacrificed as of Day
14
30 (Figure 1). Aerosol was generated by ultrasonic nebulizer in a chamber. Study was approved by
15
our institutional Animal Care and Use Committee.
16
17
2.4. Measurement of airway hyperresponsiveness
18
19
AHR was assessed by whole-body plethymography (Buxco Electronics, Troy, NY) after the last
20
OVA inhalation. Mice were placed in measured chamber and kept in a dark box. Aerosolized normal
21
saline or methacholine in increasing concentration (6.25-25 mg/ml) were nebulized via chamber inlet
22
for 3 min. Recordings and baselines were averaged for 3 min after each nebulization of variant
23
methacholine concentration. AHR was expressed as mean enhanced pause (Penh = pause × peak
24
expiratory box flow/peak inspiratory box flow), data expressed as ratio of PenhMCh and PenhNaCl.
25
6
1
2.5. Analyzing cellular composition of BALF
2
3
Mice were sacrificed by CO2 asphyxia after measuring AHR, their tracheas immediately lavaged
4
three times via catheter with 1 ml normal saline. BALF was centrifuged (400 g) at 4°C for 5 min.
5
After washing with normal saline, these cell pellets were resuspended in 1 ml normal saline. Total
6
cell number was counted by standard haemocytometer, differential cell counts performed by staining
7
with Liu’s stain solution and then counting at least 200 cells. Differentiating cell was judged by
8
standard morphological criteria.
9
10
2.6. Determination of serum antibody
11
12
Serum samples collected from hearts after sacrificing were stored at -80°C until assay. Levels of
13
serum antibodies were measured for IgE, IgA and IgG, using enzyme-linked immunosorbent assay
14
(ELISA) kits (eBioscience, San Diego, CA). OVA-specific IgE, IgG1 and IgG2a were also measured.
15
16
2.7. Preparations and cytokines expression of splenic mononuclear cells
17
18
Spleens were removed aseptically and placed individually into 5 ml complete RPMI-1640
19
medium. Single-cell suspensions were prepared by pipetting spleens into small pieces and single-cell
20
suspensions with sterile syringe. Suspension was centrifuged at 180 g for 5 min. After discarding
21
supernatant, the cell pellet was resuspended in lysis buffer (Tris-NH4Cl) to lyse erythrocytes; lysis
22
buffer was washed twice in RPMI-1640 medium, cell viability and concentration determined by a
23
hemocytometer after staining of trypan blue. Cell concentration was adjusted to 2×106 cells/ml in
24
RPMI-1640 medium, and aliquots of 200 μl suspension added to each well of a 96-well plate and
25
cultured in the presence of OVA (10 mg/ml; Sigma) for 48 h at 37°C. Cell-free supernatant fractions
7
1
were harvested and stored at -80°C until assayed. Presence of IL-12, IFN-γ, IL-4 and IL-10 in culture
2
supernatant fractions was determined by different commercial enzyme-linked immunosorbent assay
3
(ELISA) kit, as per manufacturer’s instructions.
4
5
2.8. Histologic examination
6
7
After BAL, trachea and lungs from each animal were removed immediately and fixed by 10%
8
buffered formalin. Sections of each lobe (five per animal) were stained with haematoxylin and eosin
9
for microscopic examination to rate severity of inflammation. Two pathologists (blinded) assigned
10
total inflammation score for each section of lung parenchyma: 0, normal; 1, inflammatory infiltration
11
of eosinophils, lymphocytes, and neutrophils in <25% of the entire section; 2, inflammatory infiltrate
12
in 25-50% of entire section; 3, inflammatory infiltrate >50% of entire section. Total inflammation
13
score for each animal was calculated as mean score for five lung sections.
14
15
2.9. Cellular staining procedure and cell apoptosis
16
17
Splenic monocular cells were isolated from spleen and suspended in RPMI-1640 culture
18
medium, 106 cells fixed with 4% paraformaldehyde for 30 min at 4°C, prior to staining for 5 min.
19
Stained cells were washed with phosphate buffer saline (PBS, 0.2% BSA) and permeabilized for 5
20
min on ice with 200 ul 70% ethanol. After washing, cells were washed three times with PBS
21
containing 0.2% BSA buffer, then decanted and stained either with anti-CD4 or anti-CD8 fluorescein
22
isothiocyanate (FITC)-conjugated monoclonal antibody (mAb) (eBioscience). To detect FoxP3
23
expression of CD4+ or CD8+ T, cells were permeabilized and stained with anti-FoxP3 PE conjugated
24
mAb, using standard protocol (eBioscience). Cells were analyzed by FACS scan flow-cytometer (FC
25
500, Beckman Coulter, Miami, FL).
8
1
Apoptosis of splenic mononuclear cells was studied with DNA strand breaks on the terminal
2
deoxnucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) kit (Mebsttain, Immunotch,
3
France). Briefly, 106 cells were fixed with 4% paraformaldehyde for 30 min at 4oC, washed (for 5
4
min at 1500g) with phosphated buffer saline (PBS containing 0.2% BSA) and permeabilized for 5
5
min on ice with 200 μl 70% ethanol. After washing, cells were decanted and resuspended in 30μl of
6
TUNEL labeled as a negative control. After 60 min of incubation at 37oC in a humidified atmosphere,
7
cells were washed three times with PBS containing 0.2% BSA buffer and stained with Flurescein
8
isothiocyanate (FITC)-conjugated mAb for 30 min. For simultaneous detection of apoptosis, cytokine
9
expression, and surface phenotype of a single cell level, anti-IL-4 phycoerythrin (PF) mAb, or
10
corresponding isotype control mAbs (Coulter-Immunotech, Miami, FL) were labeled at the same
11
time. After staining, cells were analyzed with a FACS scan flow cytometer (FC500, Beckman
12
Coulter Inc., USA) acquiring 10,000 events.
13
14
2.10. Statistical analysis
15
16
Data were calculated with mean values, standard deviations (mean±SD) derived from triplicate
17
trials. Significance was rated by one-way ANOVA (analysis of variance) and Duncan’s multiple range
18
tests (P<0.05) using SPSS 10.0.7 software, figure drawn using Sigma Plot 10.0.
19
20
21
22
3. Results
23
24
3.1. Pulmonary function tests
25
9
1
Mice treated with saline showed markedly higher Penh ratio after challenge with methacholine
2
at 25 mg/ml when compared with the other groups (P<0.05). At dose of methacholine tested, Penh
3
ratio was significantly lower in group of high-dose combined LAB (L. paracasei BB5 and L.
4
rhamnosus BB1) than single strain of either L. paracasei BB5 or L. rhamnosus BB1 (P<0.05) (Fig.
5
2). Meanwhile, Penh ratio was also significantly lower in high-dose combined LAB group than low
6
dose of combined LAB group (P<0.05) (Fig. 2).
7
8
9
3.2. Cell profile and cytokines of bronchoalveolar lavage fluid
In order to confirm inflammatory condition, cellular composition of BALF was analyzed, results
10
shown in Figure 3. OVA-saline treated mice of positive control group induced a marked rise in cell
11
numbers of eosinophil and macrophage (Fig. 3); naïve group had lowest total cell count. Eosinophil
12
is main inflammatory cell in BALF; mice treated with probiotic LAB exhibited significantly reduced
13
number of eosinophil and macrophage as compared with the OVA-saline treated group (P<0.05). For
14
lymphocytes, percentages of treating with high-dose combined LAB group were higher than the other
15
groups (P<0.05). For cytokines and total protein levels in BALF (Table 1). Naïve group had lowest
16
IL-10, TGF-β, and total protein levels in BALF as compared to OVA-treated groups. Those treated
17
with high-dose combined LAB mice had lower levels of TGF-β and higher levels of IL-10 than other
18
LAB treated groups, and significantly different from OVA-saline treated group (P<0.05).
19
20
3.3. Lung tissue histology
21
22
Lung tissue and trachea of the OVA-saline group showed severe inflammation, characterized by
23
hyperemia, interstitial edema, and inflammatory cell infiltration (Fig. 4a). Only mild hyperemia,
24
interstitial edema, and inflammatory cell infiltration appeared in the lung tissues and airways of the
25
high-dose combined LAB group (Fig. 4e). Inflammation was highest in the OVA-saline as compared
10
1
to groups of P-1× (Fig. 4b), R-1× (Fig. 4c), low-dose of P+R,-1× (Fig. 4d), high-dose of P+R,-3×
2
(Fig. 4d), and naïve control group (Fig. 4f). Mean inflammatory score of terminal bronchioles in the
3
high-dose combined LAB group was lower than that of the OVA-saline group (1.2±0.3 vs. 2.2±0.4
4
respectively, P<0.05). Mean inflammatory score of trachea in the high-dose combined LAB group
5
was also lower than that of the OVA-saline group (1.0±0.2 vs. 2.1±0.4 respectively, P<0.05).
6
7
3.4. Levels of serum antibody and OVA-specific antibody
8
9
Figure 5 plots total serum immunoglobulins (Ig) including IgE, IgA and IgG. Levels of serum
10
IgE treated with different LAB were lower than the OVA-saline group (Fig. 5a, P<0.05), as was level
11
of IgG (Fig. 5b). Yet levels of IgA rose in mice treated with variant LAB (Fig. 5c); those treated with
12
single strain of L. rhamnosus BB1 were higher than those treated with L. paracasei BB5 (P<0.05).
13
Serum OVA-specific immunoglobulin levels including IgE, IgG1, and IgG2a were measured (Figure
14
6). Figure 6a shows high-dose combined LAB-treatment with starkly lower OVA-specific IgE level
15
compared with OVA-saline or other LAB-treated groups (P<0.05). Serum OVA-specific IgG1 level
16
showed no significant differences among OVA-saline or other LAB-treated groups (Fig. 6b, P>0.05).
17
While IgG2a serum levels related with Th1 cell pathway, results show LAB-treated groups sharply
18
higher than the OVA-saline group (Fig. 6c, P<0.05). Combined LAB induced a stronger response of
19
IgG2a. Hence either L. paracasei BB5 or L. rhamnosus BB1 strain could inhibit Th2 cell pathway to
20
increase serum level of IgG2a and decrease serum level of specific IgE, with no effect on IgG1.
21
22
3.5. Cytokines secretion from mouse splenic mononuclear cells
23
24
To ascertain whether LAB influences secretion of IL-12, IFN-γ, IL-4 and IL-10, mice splenic
25
mononuclear cells of each group were co-cultured with OVA for 48 h. Cell viabilities of prepared
11
1
splenic cells before OVA treatment were above 95%. Figure 7 shows cytokine levels of IL-12 (Fig.
2
7a) and IFN-γ (Fig. 7b); important Th1 cytokines increased significantly in mice treated with low- or
3
high-dose combined LAB as compared with groups of naïve and OVA-saline treated mice (P<0.05).
4
IL-4 production decreased significantly in single or combined LAB-treated mice than in those treated
5
with OVA-saline (Fig. 7c, P<0.05); reduced effect of combined LAB was higher than the single LAB
6
strain. IL-10 production of splenic mononuclear cells were increased significantly after treating with
7
tested LAB as compared with naïve and OVA-saline treated mice (Fig. 7d, P<0.05).
8
9
3.6. Induced apoptosis of Th2 lymphocytes
10
11
Number of CD4+FoxP3+ T and CD8+FoxP3+ Treg cells significantly rose only in the high-dose
12
combined LAB versus OVA-saline-treated group in splenic mononuclear cells (Figs. 8a-8b), with no
13
difference emerging in CD4+FoxP3+ and CD8+FoxP3+ Treg cells between single-strain or low-dose
14
combined strain groups. Ratio of CD4+IL-4+ TUNEL cells in splenic mononuclear cells rose
15
significantly in the high-dose combined LAB-treated group, as compared to OVA-saline and other
16
LAB-treated groups (Fig. 8c).
17
18
19
20
4. Discussion
21
22
Many studies attempted to devise some novel probiotic LAB to treat allergic disease [30, 31].
23
The present study rated efficacy of single LAB and combined strains for inhibiting OVA-induced
24
allergic AHR. Results proved combination of L. paracasei BB5 or L. rhamnosus BB1 with higher
25
efficacy than treatment with single strain; it also has dose-dependent effect. Serum OVA-specific IgE
12
1
and splenic IL-4 levels were inhibited in the LAB-treated group; cytokine levels of IL-12, IL-10, and
2
IFN-γ significantly increased after stimulation with OVA on splenic cells (Figs. 6-7). This study also
3
found high-dose combined LAB had induced higher percentage of CD4+FoxP3+ and CD8+FoxP3+
4
Treg cells as well as CD4+IL-4+ apoptotic cells than the other groups (Fig. 8). Results indicate a key
5
role of combined LAB may attenuate a Th2 cell-mediated allergic immunity and induce CD4+ IL-4+
6
cell apoptosis by enhancing expression of Th1 and Treg cells.
7
Clinical study indicated supplementation of probiotics during pregnancy potentially enhancing
8
immune parameters of IFN-γ and IgA, along with immunomodulatory factors of TGF-β in breast
9
milk [55, 56]. Mixture of probiotics and prebiotics also contributed a beneficial effect to modulated
10
infant’s intestinal microbiota successfully and IgE-associated atopic dermatitis [57-59]. Feleszko et al.
11
demonstrated both Bifidobacterium lactis Bb-12 and Lactobacillus rhamnosus GG to reduce airway
12
hyperresponsiveness and percentage of inflammatory cells in BAL fluid [33]. Here we demonstrated
13
screening L. paracasei BB5 and L. rhamnosus BB1 with potential effect to relieve allergic disease by
14
immunomodulation. Our prior studies and other reports had demonstrated how diverse LAB strains
15
produced different properties, as well as immune stimulation on antigen presenting cells or peripheral
16
blood mononuclear cells (PBMC) [28, 60-62]. Tested strains in this study induce different cytokine
17
profile when co-cultured with PBMC which Der p1 of house dust-challenged or naïve mice: e.g., L.
18
paracasei BB5 induce high IFN-γ production in naïve mice, but L. rhamnosus BB1 induce high
19
IL-12 in Der p1-challenged mice. Results portend Gram-positive lactic acid bacteria producing
20
different peptidoglycans, lipoproteins, exopolysaccharide, enzyme, and cell wall. These productions
21
may induce different immune response via Toll-like receptors [63]. This study found better effect of
22
combined bacteria for regulating allergic immune response than single strain, possibly via synergistic
23
effect of different substance(s) from tested LAB strains. Other studies likewise found multistrains
24
more effective than monostrains [64, 65]. Gackowska et al. indicated LAB strains at least partially
25
dependent on their composition to regulate immune reactions. Low LAB mixture-induced IL-12 and
13
1
IFN- production and relatively high IL-10 and TNF-alpha expression may represent cellular activities
2
induced in vivo by combined action of bacterial antigens [66].
3
IL-10 plays a key role in maintenance of self-tolerance by induction of T cell with blockage of
4
CD28 T cell costimulation [45-47]. Our previous study results showed that specific-allergen induced
5
Th2 lymphocytes apoptosis in immunotherapy-treated asthmatic children or pollen-allergic patients
6
[67, 68]. In this study, increased CD4+FoxP3+ Treg cells with IL-10 production as well as IL-12 and
7
IFN-γ of Th1 cell-mediated cytokines suggest a possible mechanism of combined LAB-treatment
8
modulating Th2 to Th1 shift. Meanwhile, high-dose combined LAB treatment significantly induced
9
apoptosis of CD4+IL-4+ Th2 cells. Our prior study found that CD8+FoxP3+ Treg cells can suppress
10
CD4+ T cell proliferation by cell-contact inhibition [69]. TUNEL study revealed CD8+FoxP3 Treg
11
cells but not CD4+CD25+ Treg cells directly inducing CD4+CD45ROhigh apoptosis [51, 69]. In
12
addition, we found increased production of plasma IL-10 and TGF-β level significantly after one year
13
of immunotherapy-treating asthmatic children. During immunotherapy, both CD4+FoxP3+ and
14
CD8+FoxP3+ Treg cells increased significantly. Taken together, these in vivo human results suggest
15
increasing IL-10, TGF-β, and FoxP3+CD4/CD8+ Treg cells promotes immune tolerance. IL-10 is an
16
important cytokine to induce naïve T cell transform to Treg cells [51, 69].
17
Results indicate combination LAB treatment spawning immunomodulatory effect by inducing
18
both CD4+ and CD8+ Treg cells; it thus may be a good adjuvant for treating allergic asthma. This
19
study illustrated oral administration with high-dose combined L. paracasei BB5 and L. rhamnosus
20
BB1 ameliorating Th2 allergic AHR by boosting CD4+ Treg and CD8+ Treg cells function and
21
decreasing Th2 cytokines. Findings support the idea that combination of high-dose L. paracasei BB5
22
and L. rhamnosus BB1 as a candidate adjuvant for therapeutic intervention in allergic airway disease;
23
detailed mechanisms warrant study in the future.
24
25
14
1
2
3
4
Acknowledgments
We thank the Armed Forces Taoyuan General Hospital and National Science Council of Taiwan
for supporting this study, People Diagnosis Project 9826 and NSC 98-2313-B-039-002-MY2.
5
15
1
References
2
3
4
5
6
7
[1] Palomares O, Yaman G, Azkur AK, Akkoc T, Akdis M, Akdis CA. Role of Treg in immune
regulation of allergic diseases. Eur J Immunol 2010; 40: 1232-40.
[2] Lambrecht BN, Hammad H. The role of dendritic and epithelial cells as master regulators of
allergic airway inflammation. The Lancet 2010; 376: 835-43.
[3] Trischler J, Merkel N, Konitzer S, Muller CM, Unverzagt S, Lex C. Fractionated breath
8
condensate sampling: H2O2 concentrations of the alveolar fraction may be related to asthma
9
control in children. Respir Res 2012; 13(1): 14.
10
[4] Kashiwakura JC, Ando T, Matsumoto K, Kimura M, Kitaura J, Matho MH, Zajonc DM, Ozeki T,
11
Ra C, MacDonald SM, Siraganian RP, Broide DH, Kawakami Y, Kawakami T.
12
Histamine-releasing factor has a proinflammatory role in mouse models of asthma and allergy. J
13
Clin Invest 2012; 122(1): 218-28.
14
15
16
17
18
[5] Bjorksten B. The intrauterine and postnatal environments. J Allergy Clin Immunol 1999; 104:
1119-27.
[6] Okada H, Kuhn C, Feillet H, Bach JF. The 'hygiene hypothesis' for autoimmune and allergic
diseases: an update. Clin Exp Immunol 2010; 160(1): 1-9.
[7] Kalliomaki M, Kirjavainen P, Eerola E, Kero P, Salminen S, Isolauri E. Distinct patterns of
19
neonatal gut microflora in infants in whom atopy was and was not developing. The J Allergy
20
Clin Immunol 2001; 107: 129-34.
21
[8] Schmidt WP. Model of the epidemic of childhood atopy. Med Sci Monit 2004; 10:HY5-9.
22
[9] Arvola T, Ruuska T, Keränen J, Hyöty H, Salminen S, Isolauri E. Rectal bleeding in infancy:
23
clinical, allergological, and microbiological examination. Pediatrics 2006; 117(4): e760-68.
24
25
[10] Ghadimi D, Fölster-Holst R, de Vrese M, Winkler P, Heller KJ, Schrezenmeir J. Effects of
probiotic bacteria and their genomic DNA on TH1/TH2-cytokine production by peripheral blood
16
1
mononuclear cells (PBMCs) of healthy and allergic subjects. Immunobiology 2008; 213: 677-92.
2
[11] Wheeler JG, Shema SJ, Bogle ML, Shirrell MA, Burks AW, Pittler A. Immune and clinical
3
impact of Lactobacillus acidophilus on asthma. Ann Allergy Asthma Immunol 1997; 79(3):
4
229-33.
5
[12] Takeda S, Takeshita M, Kikuchi Y, Dashnyam B, Kawahara S, Yoshida H, Watanabe W,
6
Muguruma M, Kurokawa M.Efficacy of oral administration of heat-killed probiotics from
7
Mongolian dairy products against influenza infection in mice: alleviation of influenza infection
8
by its immunomodulatory activity through intestinal immunity. Int Immunopharmacol 2011;
9
11(12): 1976-83.
10
[13] Nagai T, Makino S, Ikegami S, Itoh H, Yamada H. Effects of oral administration of yogurt
11
fermented with Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1 and its
12
exopolysaccharides against influenza virus infection in mice. Int Immunopharmacol 2011;
13
11(12): 2246-50.
14
[14] Ghadimi D, de Vrese M, Heller KJ, Schrezenmeir J. Lactic acid bacteria enhance autophagic
15
ability of mononuclear phagocytes by increasing Th1 autophagy-promoting cytokine
16
(IFN-gamma) and nitric oxide (NO) levels and reducing Th2 autophagy-restraining cytokines
17
(IL-4 and IL-13) in response to Mycobacterium tuberculosis antigen. Int Immunopharmacol
18
2010; 10(6): 694-706.
19
[15] Kawashima T, Hayashi K, Kosaka A, Kawashima M, Igarashi T, Tsutsui H, Tsuji NM,
20
Nishimura I, Hayashi T, Obata A. Lactobacillus plantarum strain YU from fermented foods
21
activates Th1 and protective immune responses. Int Immunopharmacol 2011; 11(12): 2017-24.
22
23
24
25
[16] Dugas B, Mercenier A, Lenoir-Wijnkoop I, Amaud C, Dugas N, Postaire E. Immunity and
Probiotics. Immunology Today 1999; 20: 387-90.
[17] Sunada Y, Nakamura S, Kamei C. Effect of Lactobacillus acidophilus strain L-55 on the
development of atopic dermatitis-like skin lesions in NC/Nga mice. Int Immunopharmacol 2008;
17
1
2
3
4
5
6
7
8
9
10
11
8: 1761-66.
[18] Hsu CH, Wang MF, Lin HC, Wang YY. Treatment of perennial allergic rhinitis with lactic acid
bacteria. Pediatr Allergy Immunol 2004; 15: 152-58.
[19] Zoetendal EG, Rajilic-Stojanovic M, de Vos WM. High-throughput diversity and functionality
analysis of the gastrointestinal tract microbiota. Gut 2008; 57: 1605-15.
[20] Bjorksten B. Genetic and environmental risk factors for the development of food allergy. Curr
Opin Allergy Clin Immunol 2005; 5: 249-53.
[21] Zhang LL, Chen X, Zheng PY, Luo Y, Lu GF, Liu ZQ, Huang H, Yang PC. Oral
Bifidobacterium modulates intestinal immune inflammation in mice with food allergy. J
Gastroenterol Hepatol 2010; 25: 928-34.
[22] Smits HH, Engering A, van der Kleij D, de Jong EC, Schipper K, van Capel TM, Zaat BA,
12
Yazdanbakhsh M, Wierenga EA, van Kooyk Y, Kapsenberg ML. Selective probiotic bacteria
13
induce IL-10-producing regulatory T cells in vitro by modulating dendritic cell function through
14
dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin. The J Allergy Clin
15
Immunol 2005; 115: 1260-67.
16
[23] Donkor ON, Ravikumar M, Proudfoot O, Day SL, Apostolopoulos V, Paukovics G, Vasiljevic T,
17
Nutt SL, Gill H. Cytokine profile and induction of T helper type 17 and regulatory T cells by
18
human peripheral mononuclear cells after microbial exposure. Clin Exp Immunol 2012; 167(2):
19
282-95.
20
[24] Pozo-Rubio T, Mujico JR, Marcos A, Puertollano E, Nadal I, Sanz Y, Nova E.
21
Immunostimulatory effect of faecal Bifidobacterium species of breast-fed and formula-fed
22
infants in a peripheral blood mononuclear cell/Caco-2 co-culture system. Br J Nutr 2011; 106(8):
23
1216-23.
24
[25] Niers LE, Timmerman HM, Rijkers GT, van Bleek GM, van Uden NO, Knol EF, Kapsenberg
25
ML, Kimpen JL, Hoekstra MO. Identification of strong interleukin-10 inducing lactic acid
18
1
2
bacteria which down-regulate T helper type 2 cytokines. Clin Exp Allergy 2005; 35: 1481-89.
[26] Fink LN, Zeuthen LH, Ferlazzo G, Frokiaer H. Human antigen-presenting cells respond
3
differently to gut-derived probiotic bacteria but mediate similar strain-dependent NK and T cell
4
activation. FEMS Immunol Med Microbiol 2007; 51: 535-46.
5
6
7
[27] Meijerink M, Wells JM. Probiotic modulation of dendritic cells and T cell responses in the
intestine. Benef Microbes 2010; 1(4): 317-26.
[28] Lin WH, Wu CR, Fang TJ, Lee MS, Lin KL, Chen HC, Huang SY, Hseu YC. Adherent
8
properties and macrophage activation ability of 3 strains of lactic acid bacteria. J Food Sci 2011;
9
76(1): M1-7.
10
[29] Morita H, He F, Fuse T, Ouwehand AC, Hashimoto H, Hosoda M. Adhesion of lactic acid
11
bacteria to Caco-2 cells and their effect on cytokine secretion. Microbiol Immunol 2002; 46:
12
293-97.
13
14
[30] Damaskos D, Kolios G. Probiotics and prebiotics in inflammatory bowel disease: microflora 'on
the scope'. Br J Clin Pharmacol 2008; 65(4): 453-67.
15
[31] Walker WA. Mechanisms of action of probiotics. Clin Infect Dis 2008; 46 Suppl 2: S87-91.
16
[32] Blumer N, Sel S, Virna S, Patrascan CC , Zimmermann S, Herz U , Renz H , Garn H. Perinatal
17
maternal application of Lactobacillus rhamnosus GG suppresses allergic airway inflammation in
18
mouse offspring. Clin Exp Allergy 2007; 37: 348-57.
19
[33] Feleszko W, Jaworska J, Rha R.D, Steinhausen S, Avagyan A, Jaudszus A, Ahrensn B,
20
Groneberg D.A, Wahn U, Hamelmann E. Probiotic-induced suppression of allergic sensitization
21
and airway inflammation is associated with an increase of T regulatory-dependent mechanisms
22
in a murine model of asthma. Clin Exp Allergy 2007; 37: 498-505.
23
24
25
[34] Boyle RJ, Tang ML. The role of probiotics in the management of allergic disease. Clin Exp
Allergy 2006; 36: 568-76.
[35] Isolauri E, Arvola T, Sutas Y, Moilanen E, Salminen S. Probiotics in the management of atopic
19
1
2
eczema. Clin Exp Allergy 2000; 30: 1604-10.
[36] Liu YW, Su YW, Ong WK, Cheng TH, Tsai YC. Oral administration of Lactobacillus plantarum
3
K68 ameliorates DSS-induced ulcerative colitis in BALB/c mice via the anti-inflammatory and
4
immunomodulatory activities. Int Immunopharmacol 2011; 11(12): 2159-66.
5
6
[37] Girardin M, Seidman EG. Indications for the use of probiotics in gastrointestinal diseases. Dig
Dis 2011; 29(6): 574a-87.
7
[38] Hammer HF. Gut microbiota and inflammatory bowel disease. Dig Dis 2011; 29(6): 550-53.
8
[39] Espeche Turbay MB, de Moreno de Leblanc A, Perdigón G, Savoy de Giori G, Hebert EM.
9
β-Casein hydrolysate generated by the cell envelope-associated proteinase of Lactobacillus
10
delbrueckii ssp. lactis CRL 581 protects against trinitrobenzene sulfonic acid-induced colitis in
11
mice. J Dairy Sci 2012; 95(3): 1108-18.
12
[40] Zakostelska Z, Kverka M, Klimesova K, Rossmann P, Mrazek J, Kopecny J, Hornova M,
13
Srutkova D, Hudcovic T, Ridl J, Tlaskalova-Hogenova H. Lysate of probiotic Lactobacillus casei
14
DN-114 001 ameliorates colitis by strengthening the gut barrier function and changing the gut
15
microenvironment. PLoS One 2011; 6(11): e27961.
16
[41] Duary RK, Bhausaheb MA, Batish VK, Grover S. Anti-inflammatory and immunomodulatory
17
efficacy of indigenous probiotic Lactobacillus plantarum Lp91 in colitis mouse model.
18
Molecular Biology Reports 2012; 39(4): 4765-75.
19
[42] Ferreira RB, Gill N, Willing BP, Antunes LC, Russell SL, Croxen MA, Finlay BB. The intestinal
20
microbiota plays a role in Salmonella-induced colitis independent of pathogen colonization.
21
PLoS One 2011; 6(5): e20338.
22
[43] Peng GC, Hsu CH. The efficacy and safety of heat-killed Lactobacillus paracasei for treatment
23
of perennial allergic rhinitis induced by house-dust mite. Pediatr Allergy Immunol 2005; 16:
24
433-38.
25
[44] Xiao JZ, Kondo S, Yanagisawa N, Takahashi N, Odamaki T, Iwabuchi N. Effect of probiotic
20
1
Bifidobacterium longum BB536 in relieving clinical symptoms and modulating plasma cytokine
2
levels of Japanese cedar pollinosis during the pollen season: A randomized double-blind,
3
placebo-controlled trial. J Invest Allergol Clin Immunol 2006; 16: 86-93.
4
[45] Jutel M, Akdis M, Budak F, Aebischer-Casaulta C, Kurt Blaser M.W, Akdis C.A. IL-10 and
5
TGF-β cooperate in the regulatory T cell response to mucosal allergens in normal immunity and
6
specific immunotherapy. Eur J Immunol 2003; 33: 1205-14
7
8
9
10
11
[46] Tran DQ. TGF-β: the sword, the wand, and the shield of FOXP3(+) regulatory T cells. J Mol
Cell Biol 2012 Feb; 4(1): 29-37.
[47] Duan W, So T, Mehta AK, Choi H, Croft M. Inducible CD4+LAP+Foxp3- regulatory T cells
suppress allergic inflammation. J Immunol 2011; 187(12): 6499-507.
[48] Caramalho I, Lopes-Carvalho T, Osler D , Zelenay S, Haury M , Demengeot J. Regulatory T
12
cells selectively express toll-like receptors and are activated by lipopolysaccharide. J Exp Med
13
2003; 197: 403-11.
14
[49] Chen Q, Davidson TS, Huter EN, Shevach EM. Engagement of TLR2 does not reverse the
15
suppressor function of mouse regulatory T cells, but promotes their survival. J Immunol 2009;
16
183(7): 4458-66.
17
18
[50] Hsieh CS, Lee HM, Lio CW. Selection of regulatory T cells in the thymus. Nat Rev Immunol
2012; 12(3): 157-67.
19
[51] Tsai YG, Chiou YL, Chien JW, Wu HP, Lin CY. Induction of IL-10+CD4+CD25+ regulatory T
20
cells with decreased NF-κB expression during immunotherapy. Pediatr Allergy Immunol 2010;
21
21: e166-73.
22
[52] Hong HJ, Kim E, Cho D, Kim T.S. Differential suppression of heat-killed lactobacilli isolated
23
from kimchi, a Korean traditional food, on airway hyper-responsiveness in mice. J Clin Immunol
24
2010; 30: 449-58.
25
[53] Ahui ML, Champy P, Ramadan A, Pham Van L, Araujo L, Brou André K, Diem S, Damotte D,
21
1
Kati-Coulibaly S, Offoumou MA, Dy M, Thieblemont N, Herbelin A. Ginger prevents
2
Th2-mediated immune responses in a mouse model of airway inflammation. Int
3
Immunopharmacol 2008; 8(12): 1626-32.
4
[54] Bao ZS, Hong L, Guan Y, Dong XW, Zheng HS, Tan GL, Xie QM. Inhibition of airway
5
inflammation, hyperresponsiveness and remodeling by soy isoflavone in a murine model of
6
allergic asthma. Int Immunopharmacol 2011; 11(8): 899-906.
7
[55] Prescott SL, Wickens K, Westcott L, Jung W, Currie H, Black PN, Stanley TV, Mitchell EA,
8
Fitzharris P, Siebers R, Wu L, Crane J. Probiotic Study Group. Supplementation with
9
Lactobacillus rhamnosus or Bifidobacterium lactis probiotics in pregnancy increases cord blood
10
interferon-c and breast milk transforming growth factor-b and immunoglobin A detection. Clin
11
Exp Allergy 2008; 38: 1606-14.
12
[56] Prokesová L, Lodinová-Zádníková R, Zizka J, Kocourková I, Novotná O, Petrásková P, Sterzl I.
13
Cytokine levels in healthy and allergic mothers and their children during the first year of life.
14
Pediatr Allergy Immunol 2006; 17(3): 175-83.
15
[57] Van Aalderen WM, Heymans HS, Henk Sillevis Smitt J, Nauta AJ, Knippels LM, Ben Amor K,
16
Sprikkelman AB; Synbad Study Group. Synbiotics prevent asthma-like symptoms in infants with
17
atopic dermatitis. Allergy 2011; 66(2): 170-77.
18
19
20
[58] West CE, Hammarström ML, Hernell O. Probiotics during weaning reduce the incidence of
eczema. Pediatr Allergy Immunol 2009; 20(5): 430-37.
[59] Segawa S, Hayashi A, Nakakita Y, Kaneda H, Watari J, Yasui H. Oral administration of
21
heat-killed Lactobacillus brevis SBC8803 ameliorates the development of dermatitis and inhibits
22
immunoglobulin E production in atopic dermatitis model NC/Nga mice. Biol Pharm Bull 2008;
23
31: 884-89.
24
[60] Donkor ON, Ravikumar M, Proudfoot O, Day SL, Apostolopoulos V, Paukovics G, Vasiljevic T,
25
Nutt SL, Gill H. Cytokine profile and induction of T helper type 17 and regulatory T cells by
22
1
human peripheral mononuclear cells after microbial exposure. Clin Exp Immunol 2012; 167:
2
282-95.
3
[61] de Roock S, van Elk M, van Dijk ME, Timmerman HM, Rijkers GT, Prakken BJ, Hoekstra MO,
4
de Kleer IM. Lactic acid bacteria differ in their ability to induce functional regulatory T cells in
5
humans. Clin Exp Allergy 2010; 40: 103-10.
6
[62] Hua MC, Lin TY, Lai MW, Kong MS, Chang HJ, Chen CC. Probiotic Bio-Three induces Th1
7
and anti-inflammatory effects in PBMC and dendritic cells. World J Gastroenterol. 2010 Jul 28;
8
16(28): 3529-40.
9
[63] Nada M, Ohnishi H, Tochio H, Kato Z, Kimura T, Yamamoto T, Kamatari YO, Tsutsumi N,
10
Shirakawa M, Kondo N. Molecular analysis of the binding mode of Toll/interleukin-1 receptor
11
(TIR) domain proteins during TLR2 signaling. Mol Immunol 2012; 52: 108-16.
12
[64]Lin WH, Yu B, Lin CK, Hwang WZ, Tsen HY. Immune effect of heat-killed multistrain of
13
Lactobacillus acidophilus against Salmonella typhimurium invasion to mice. J Appl Microbiol
14
2007; 102: 22-31.
15
[65]Timmerman HM, Koning CJ, Mulder L, Rombouts FM, Beynen AC. Monostrain, multistrain and
16
multispecies probiotics-A comparison of functionality and efficacy. Int J Food Microbiol 2004;
17
96: 219-33.
18
[66] Gackowska L, Michalkiewicz J, Krotkiewski M, Helmin-Basa A, Kubiszewska I,
19
Dzierzanowska D. Combined effect of different lactic acid bacteria strains on the mode of
20
cytokines pattern expression in human peripheral blood mononuclear cells. Physiol Pharmacol
21
2006; 57 Suppl 9: 13-21.
22
[67] Guerra F, Carracedo J, Solanan-Lara R, Sanchez-Guuo P, Ramirez R. Th2 lymphocytes from
23
atopic patients treated with immunotherapy undergo rapid apoptosis after culture with specific
24
allergens. The J Allergy Clin Immunol 2002; 109: 263.
25
[68] Tsai YG, Chien JW, Chen WL, Shieh JJ, Lin CY. Induced apoptosis of TH2 lymphocytes in
23
1
asthmatic children treated with Dermatophagoides pteronyssinus immunotherapy. Pediatr
2
Allergy Immunol 2005; 16(7): 602-08.
3
[69] Tsai YG, Tsai YG, Yang KD, Niu DM, Chien JW, Lin CY. TLR2 agonists enhance CD8+Foxp3+
4
regulatory T cells and suppress Th2 immune responses during allergen immunotherapy. J
5
Immunol 2010; 184(12): 7229-37.
6
24
1
Figure legends
2
3
Fig. 1. Asthma induced protocol: Mice were fed orally with different samples daily from Day 0 to 28.
4
On Days 14 and 21, mice were immunized intraperitoneally (i.p) with 50 μg OVA. On Days
5
27-28, mice were challenged with OVA aerosol inhalation for 10 minutes; then the airway
6
hyper-responsiveness was measured after aerosolized methacholine challenge on Day 29.
7
Mice were sacrificed on Day 30.
8
9
Fig. 2. Effects on airway hyperresponsiveness after daily oral administration of single or combined
10
LAB from Days 0 to 28, airway Penh ratio estimated as lung function index after stimulation
11
with aerosolized methacholine on mice of OVA-induced allergic asthma, values expressed as
12
mean±SD (n=8). Letters represent significant difference (P<0.05).
13
14
15
Fig. 3. Cell profile of bronchoalveolar lavage fluid. Values are expressed as mean±SD (n=8). Letters
represent significantly different cells (P<0.05).
16
17
Fig. 4. Histopathology of airway inflammation. OVA-saline group showed severe inflammation of
18
lung tissue and trachea, characterized by hyperemia, interstitial edema and inflammatory cell
19
infiltration. Inflammation was highest in the OVA-saline group (4a) versus P-1 (4b), R-1 (4c),
20
P+R,-1× (4d), P+R,-3× (4e), and naïve control (4f). Hematoxylin and eosin stain 200×.
21
22
23
Fig. 5. Levels of nonspecific IgE (5a), IgG (5b) and IgA (5c) in mice sera, values expressed as mean
±SD (n=8). Different letters represents significantly difference (P<0.05).
24
25
Fig. 6. Levels of OVA-specific IgE (6a), IgG1 (6b) and IgG2a (6c) in mice sera, values expressed as
25
1
mean±SD (n=8). Different letters represent significant differences (P<0.05).
2
Fig. 7. Cytokine secretion of IL-12 (7a), IFN-γ (7b), IL-4 (7c) and IL-10 (7d) from mice spleen
3
mononuclear cells after treating with OVA, values expressed as mean±SD (n=8). Different
4
letters represent significant differences (P<0.05).
5
6
Fig. 8. Percentage of splenic monomclear cells with positive staining for CD4+FoxP3+ lymphocytes
7
(8a), CD8+FoxP3+ lymphocytes (8b) and CD4+IL-4+ TUNEL (8c), values expressed as mean±
8
SD (n=8). Different letters represents significantly difference (P<0.05).
9
26
1
2
Methacholine
challenge
3
OVA i.p
4
OVA i.p
1st & 2nd
inhalation
5
Sacrific
e
6
7
Day
0
8
14
21
Oral feeding different samples for 28 days
9
10
11
Fig. 1
12
27
27 28
29
30
1
2
6
Naive
OVA
P-1x
R-1x
P+R,-1x
P+R,-3x
Penh ratio
5
a
4
a
b
3
c
a
2
Saline
5
c
cd
b
c
c
c
c
1
6.25
d
d
12.5
Methacholine (mg/ml)
3
4
b
Fig. 2
6
28
c
c
d
25
1
2
60
a
Naive
OVA
P-1x
R-1x
P+R,-1x
P+R,-3x
Cell number (x104)
50
40
b
b
b
b
30
a
20
a
b
c
10
cc
d
bb
c
b
b
a a a aa
c
0
a
Macrophage Eosinophils Lymphocytes Basophils
3
4
5
Fig. 3
6
29
1
2
Fig. 4a
Fig. 4b
Fig. 4c
Fig. 4d
Fig. 4e
Fig. 4f
3
4
5
6
7
8
9
30
600
a
500
IgE (ng/ml)
400
300
b
200
b
b
b
100
c
0
1
2
Naive
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 5a
800
a
b
IgG (ng/ml)
600
b
b
b
b
400
200
0
3
4
Naive
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 5b
1600
a
1400
a
IgA (ng/ml)
1200
b
1000
800
600
a
c
d
400
200
0
5
6
Naive
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 5c
7
31
1
a
200
ab
bc
OVA-IgE (ng/ml)
c
150
d
100
50
ND
0
2
3
Naive
OVA
P-1x
a
a
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 6a
250
a
a
a
OVA-IgG1 (ng/ml)
200
150
100
50
ND
0
4
5
Naive
R-1x
P+R,-1x P+R,-3x
Fig. 6b
180
a
160
OVA-IgG2a (ng/ml)
140
b
120
c
c
100
80
d
60
40
20
ND
0
6
7
Naive
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 6c
8
32
1
600
a
200
b
150
IFN-(pg/ml)
IL-12 (pg/ml)
500
100
bc
50
a
400
b
300
c
200
d
c
c
100
c
e
f
0
0
Naive
2
3
OVA
P-1x
R-1x
Naive
P+R,-1x P+R,-3x
Fig. 7a
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 7b
40
30
a
a
25
b
c
cd
d
20
IL-10 (pg/ml)
IL-4 (pg/ml)
30
b
20
b
10
c
10
c
5
e
0
4
5
b
15
0
Naive
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Naive
Fig. 7c
Fig. 7d
6
33
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
35
a
CD4+FoxP3+ (%)
30
25
bc
b
bc
20
c
15
10
d
5
0
1
2
Naive
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 8a
18
a
16
CD8+FoxP3+ (%)
14
12
bc
10
bc
b
c
8
6
4
d
2
0
3
4
Naive
OVA
P-1x
R-1x
P+R,-1x P+R,-3x
Fig. 8b
12
a
CD4+IL-4+ TUNEL (%)
10
b
8
d
d
Naive
OVA
c
c
P-1x
R-1x
6
4
2
0
5
6
P+R,-1x P+R,-3x
Fig. 8c
34
1
Table 1
2
IL-10, TGF-β, and total protein levels in bronchoalveolar lavage fluid
Naïve
OVA
P-1×
R-1×
P+R,-1×
P+R,-3×
IL-10 (pg/ml)
9.8±2.3 c
32.5±6.4 b
33.2±7.3 ab
38.1±5.7 ab
41.2±6.3 ab
46.5±5.4 a
TGF-β (pg/ml)
5.2±1.4 c
18.4±4.6 a
17.6±5.2 a
16.3±4.8 a
15.6±4.2 a
12.3±3.9 b
Protein (μg/ml) 96.5±20.4 b 312.4±62.3 a 321.2±58.4 a 343.5±51.4 a 351.2±53.4 a 381.3±55.8 a
3
Each value in the table represents mean value ± SD (n=8). Values in the same row followed by
4
different letters differ significantly (P<0.05) for treatment groups.
35
Download