Introduction: Asthma is an inflammatory reaction mediated by type 2 helper T (Th2) cells and is known to increase eosinophil levels. Our previous study showed that stress-related asthma can cause neutrophilic and eosinophilic airway inflammation by suppressing immune tolerance. However, the mechanism of stress-induced neutrophilic and eosinophilic airway inflammation remains unclear. Therefore, to elucidate the cause of neutrophilic and eosinophilic inflammation, we investigated the immune response during the induction of airway inflammation. In addition, we focused on the relationship between immune response modulation immediately after stress exposure and the development of airway inflammation. Methods: Asthmatic mice were induced by three phases using female BALB/c mice. During the first phase, the mice were made to inhale ovalbumin (OVA) to induce immune tolerance before sensitization. Some mice were exposed to restraint stress during the induction of immune tolerance. In the second phase, the mice were sensitized with OVA/alum intraperitoneal injections. In the final phase, onset of asthma was induced through OVA exposure. Asthma development was evaluated based on airway inflammation and T-cell differentiation. Microarray and qPCR analyses were used to enumerate candidate factors to investigate the starting point of immunological modification immediately after stress exposure. Furthermore, we focused on interleukin-1β (IL-1β), which initiates these immune modifications, and performed experiments using its receptor blocker interleukin-1 receptor antagonist (IL-1RA). Results: Stress exposure during immune tolerance induction increased eosinophil and neutrophil airway infiltration. This inflammation was associated with decreased T regulatory cell levels and increased Th2 and Th17 levels in bronchial lymph node cells. Microarray and qPCR analyses showed that the initiation of Th17 differentiation might be triggered by stress exposure during tolerance induction. IL-1RA administration during stress exposure suppressed neutrophilic and eosinophilic airway inflammation via Th17 reduction and Treg increase. Conclusions: Our results show that psychological stress causes both eosinophilic and neutrophilic inflammatory responses due to the breakdown of immune tolerance. Furthermore, stress-induced inflammation can be abolished using IL-1RA.

Bronchial asthma is a disease in which the mucous membrane thickens due to chronic inflammation of the bronchi and narrowing of the airways, leading to wheezing and dyspnea. In 2017, there were 273 million people with asthma (prevalence rate 3.57%), and the number of deaths due to asthma was 490,000 (mortality rate 0.006%) [1]. Aspirin- and exercise-induced asthma are two common phenotypes of asthma with distinct clinical, physiological, and morphological characteristics. Asthma phenotypes are challenging to treat due to the heterogeneity of the disease manifestations [2]. To facilitate treatment, it is necessary to classify asthma into endotypes based on the underlying molecular pathology [2].

In our previous studies, we reported that exposure to psychological stress during the induction phase of respiratory tolerance, but not during the sensitization phase, promoted the development of type 2 helper T (Th2)-biased sensitization and, consequently, induced asthmatic airway responses to inhaled allergens. In contrast, restraint stress exposure did not affect sensitization and allergic airway inflammation [3‒5]. Tolerance to ovalbumin (OVA) before sensitization in asthmatic mice increases the levels of T regulatory (Treg) cells [6, 7]. Treg cells maintain immunological unresponsiveness to self-antigens and suppress excessive immune responses that are detrimental to the host [8]. Previously, we reported that stress disrupts immune tolerance [5, 9]. The level of Treg cells, confirmed by a transcription factor Foxp3, was increased in tolerized mice compared to stressed/tolerized mice [5, 10, 11].

In addition, we previously reported elevated neutrophils in psychologically stressed mice with a compromised immune tolerance [5, 9]. Th17 cells are involved in the neutrophil immune responses. For naive T cells to differentiate into Th17 cells, cytokines, such as IL-1β, and transcription factors, such as retinoid-related orphan receptor γt (RORγt), are required [12, 13]. Furthermore, it has been pointed out that Th17 response enhancement through induction of IL-1β production is involved in immune modulation by stress exposure. Exacerbation of symptoms by stress exposure IL1-β production has been implicated in multiple sclerosis [14, 15]. These findings suggest that psychological stress has the potential to activate the IL-17 immune response. Therefore, IL-1β-associated Th17 cell immune responses may play a role in the breakdown of immune tolerance due to stress exposure.

Stress is inevitable in modern-day life. However, suppression of stress-induced effects could prevent the exacerbation of asthma [3]. Therefore, in this study, we focused on the stress-IL-1β-Th17 axis to evaluate the impact of allergen-induced pre-sensitization stress on the development of asthma. Furthermore, we examined the potential of an interleukin-1 receptor antagonist (IL-1RA) to suppress stress-related asthma.

Mice

Specific pathogen-free female BALB/c mice were purchased from CLEA Japan (Tokyo, Japan). Mice were housed under a 12-h light/dark cycle at a constant temperature of 23 ± 1°C. Sterilized food and water were provided ad libitum. All experiments described below were approved by the Committee of Animal Experiments at Tohoku Medical and Pharmaceutical University (approval numbers: 16002-cn, 17004-cn, A2040, A2207). All experiments were performed in compliance with relevant institutional guidelines.

Protocols for Tolerization, Sensitization, Antigen Challenge, and Stress Exposure

Seven-week-old mice were sensitized and exposed to inhalation antigens as described previously [5] (shown in Fig. 1a). Briefly, respiratory tolerance was induced by two instances of inhalation of chicken (grade V; Sigma-Aldrich, St. Louis, MO, USA) without aluminum hydroxide on days 6 and -3. In the tolerance induction phase for the first 30 min of stress exposure time (described below), aerosolized OVA (5 mg/mL in saline) flowed through the chambers for OVA exposure. For the control group, saline alone was inhaled through the chambers (asthmatic mice). To induce restraint stress, each mouse was placed in a 50-mL conical centrifuge tube with multiple ventilation holes [5]. This restraint allowed the mice to rotate from a supine to a prone position but not turn their heads toward their tails or consume food and water. Restraint stress is generally considered to be able to induce psychological stress in animals [16]. During the tolerance phase, stressed/tolerized and stressed mice in 50-mL conical centrifuge tubes were left in plastic chambers for 6 h for 6 consecutive days (−7 to −2) at the same time each day. Non-stressed mice were deprived of food and water during the period when the stressed mice were exposed to stress. Then, mice were sensitized via intraperitoneal injection of OVA (8 μg/mouse) adsorbed onto aluminum hydroxide (FUJIFILM Wako Pure Chemical Corporation, Osaka, Japan; 4 mg/mouse) on day zero and 5. After the challenge, bronchoalveolar lavage (BAL) was performed [17].

Fig. 1.

Effects of immune tolerance and stress on allergic airway responses. a Schematic representation of the protocols used for tolerization, stress exposure, sensitization, and antigen challenge. b BALF was obtained from mice of each group 5 days after the last OVA challenge (day 22). Total cells, eosinophils, and neutrophils were counted. c, d Effects of stress on OVA-specific IgE (c) and IgG1 (d) in serum. Sera were obtained from mice of each treatment group 5 days after the last OVA challenge (day 22). OVA-specific IgE and IgG1 levels in serum were measured by ELISA. Data are expressed as the mean ± SD (n = 4–8 from 3 independent experiments). *p < 0.05, **p < 0.01, and ***p < 0.001.

Fig. 1.

Effects of immune tolerance and stress on allergic airway responses. a Schematic representation of the protocols used for tolerization, stress exposure, sensitization, and antigen challenge. b BALF was obtained from mice of each group 5 days after the last OVA challenge (day 22). Total cells, eosinophils, and neutrophils were counted. c, d Effects of stress on OVA-specific IgE (c) and IgG1 (d) in serum. Sera were obtained from mice of each treatment group 5 days after the last OVA challenge (day 22). OVA-specific IgE and IgG1 levels in serum were measured by ELISA. Data are expressed as the mean ± SD (n = 4–8 from 3 independent experiments). *p < 0.05, **p < 0.01, and ***p < 0.001.

Close modal

Measurement of Eosinophils and Neutrophils

BALB/c mice were divided into four groups: asthmatic mice, stressed mice, tolerized mice, and stressed/tolerized mice. Bronchoalveolar lavage fluid (BALF) was obtained from mice in each treatment group 5 days after the last OVA challenge (day 22). BALF was recovered by injecting 0.25 mL of 4°C cold phosphate-buffered saline (PBS) twice to wash the lungs, and approximately 0.4 mL of the recovered PBS was pooled and obtained from each mouse. BAL cells (2 × 105) were stained with DiffQuik solution (Sysmex Co., Kobe, Japan), and cell differential percentage was determined by counting a minimum of 200 cells via light microscopy [5].

OVA-Specific IgE and IgG1 Measurement

The concentrations of OVA-specific IgE and IgG1 in serum collected on day 22 were measured by ELISA, as described previously [17]. The sensitivity of detection was 1.9 × 10−2 EU/mL for IgE and 1.9 × 10−2 EU/mL for IgG1.

Preparation of the Lung Homogenate

The entire lungs of mice were homogenized in chilled 0.1% Triton-X PBS with 1% protease inhibitor (Sigma-Aldrich). After centrifugation at 15,000×g for 15 min at 4°C, the supernatants were harvested and stored at −80°C until cytokine analysis [18].

Measurement of Cytokine Concentration

The IL-1β and IL-17A levels were measured using ELISA kits (R&D Systems, Minneapolis, MN, USA), and the detection limits were 2.31 pg/mL and 5 pg/mL, respectively. Total protein levels in lung homogenates were measured using a detergent-compatible protein assay kit (Bio-Rad Laboratories, Hercules, CA, USA). The cytokine concentration in the lungs was adjusted for the protein level in each lung [18].

Preparation of RNA and Microarray Hybridization

Bronchial lymph nodes (BLN) were collected on day −2 from the stressed/tolerized mice and tolerized mice. RNA was extracted using the RealiaPrep RNA Cell Miniprep System (Promega, Madison, WI, USA) according to the manufacturer’s protocol.

Microarray Data Analysis

Microarray hybridization was performed by Kurabo Industries, Ltd. RNA quality was first assessed using an Agilent 2100 bioanalyzer, and only samples with RNA integrity number values >8 were subsequently used. A mouse Clariom S microarray (Thermo Fisher Scientific, Waltham, MA, USA) was used to analyze only the fully annotated genes. The Clariom S Array can analyze more than 22,100 genes in mice, and the gene transcript levels were determined using the Microarray Analysis Suite Software Transcriptome Viewer (KURABO Industries Ltd.).

Reverse Transcription-Quantitative Polymerase Chain Reaction

Total RNA was extracted from BLN. RNA was extracted as previously described. cDNA was synthesized using the PrimeScript RT reagent Kit (Takara Bio Inc. Kusatsu, Shiga, Japan) following the manufacturer’s protocol. PCR was performed using Power SYBR Green Master Mix and the Step One Real-Time PCR System (Thermo Fisher Scientific). HPRT expression was used as the internal control. The following primers synthesized by Eurofines Genomics (Tokyo, Japan) were used: Hprt forward 5′-CTT CCT CCT CAG ACC GCT TT-3′, reverse 5′-CAT CATC GCT AAT CAC GAC GC-3′; Rorc forward 5′-GAC CCA CAC CTC ACA AAT TGA-3′, reverse 5′-AGT AGG CCA CAT TAC ACT GCT-3′; Icos forward 5′- ATG AAG CCG TAC TTC TGC CG-3′, reverse 5′-AGT AGG CCA CAT TAC ACT GCT-3′. qPCR analysis cycling conditions were as follows: an initial denaturing step at 95°C for 10 min, followed by 40 cycles of a denaturation step (95°C for 10 s) and an annealing/extension step (60°C for 30 s). Fold changes in gene expression were determined using standard curve method.

Flow Cytometric Analysis

BLN was collected on day 20 from the mice in the four groups: asthma, stress, tolerized, and stress/tolerized groups. The BLN cells were preincubated with anti-CD16/CD32 (FC gamma III/II receptor; BD Biosciences, Franklin Lakes, NJ, USA) to reduce the nonspecific binding of the subsequent antibodies. Dead cells were excluded using the LIVE/DEAD Fixable Blue Dead Cell Stain Kit (Thermo Fisher Scientific). The cells were stained for surface antigens with anti-CD3ε-PerCP-Cy5.5 (clone 145-2C11; Miltenyi Biotec, Bergisch Gladbach, Germany), anti-CD4-AF700 (clone GK1.5; BD Biosciences), or isotype control antibodies. For intracellular staining, cells were stimulated with phorbol 12-myristate 13-acetate (50 ng/mL; Sigma-Aldrich), ionomycin (1,000 ng/mL; Sigma-Aldrich), and monensin (2 μm; BioLegend, San Diego, CA, USA) for 4 h before surface antigen staining. After fixation and permeabilization, the cells were incubated with anti-Foxp3-PE-Cy7 (clone FJK-16s; Thermo Fisher Scientific), anti-IL-17A-APC-Cy7 (clone.TC11-18H10.1; BioLegend), anti-IL4-PE-CF594 (clone 11B11; BioLegend), or an isotype control antibody. We considered CD3+CD4+Foxp3+ cells as Treg cells, CD3+CD4+IL-4+ cells as Th2 cells, and CD3+CD4+IL-17a+ cells as Th17 cells. The cells were counted on a FACSAria II flow cytometer (BD Biosciences), and the analyses were performed using the FACSDiva software (BD Biosciences).

Administration of IL-1 Receptor Antagonist

IL-1RA (140 μg/kg i.p.) (BioLegend) was administered for 60 min before stress exposure between day −7 and −2.

Statistical Analysis

Data are expressed as the mean ± standard deviation from multiple independent experiments (as indicated by n values). Significant differences between the two groups were determined using the nonparametric Mann-Whitney U test. The analyses were performed using Prism 6 (GraphPad Software, San Diego, CA, USA). Statistical significance was set at p < 0.05.

Measurement of Eosinophils and Neutrophils in BALF and IgE and IgG1 in Serum

We investigated the mechanism underlying psychological stress in the development of neutrophilic and eosinophilic airway inflammations and the induction of IgE and IgG1 synthesis. First, we evaluated the effect of psychological stress on airway inflammation after antigen challenge in the four groups of mice (shown in Fig. 1b). Airway inflammation levels were analyzed by determining eosinophil and neutrophil infiltration in BALF. On day 22, BALF was collected from the mice in the four groups. The number of eosinophils in the tolerized mice was significantly lower than in the other groups. The number of neutrophils in the stress/tolerized group was significantly higher than that in the other groups. Next, we evaluated the effect of psychological stress on OVA-specific IgE and IgG1 synthesis after antigen challenge in the four groups of mice (Fig. 1c, d). As expected, tolerized mice showed significantly lower OVA-specific IgE and IgG1 levels in serum than asthmatic mice. In contrast, The IgE and IgG1 levels were significantly higher in stressed/tolerized mice than in tolerized mice. However, no significant differences were observed in airway responses or IgE and levels between asthmatic and stressed/asthmatic mice. This likely reflects disruption of immune tolerance by stress exposure. Having shown that neutrophil infiltration was detected in stress/tolerized mice, we investigated whether neutrophil infiltration was dependent on IL-17A expression in the lungs after stress exposure. IL-17A levels were measured in the lungs of the mice in the four groups on day 20 (shown in Fig. 2). The mice in the stress/tolerized group showed increased IL-17A levels. This result suggests the increased activation of Th17 cells in the lungs of stress/tolerized mice.

Fig. 2.

Expression level of IL-17A due to psychological stress. IL-17A levels in the lung were determined via ELISA. The data from three independent experiments for each group are expressed as the mean ± SD (n = 5–8 from 3 independent experiments). *p < 0.05.

Fig. 2.

Expression level of IL-17A due to psychological stress. IL-17A levels in the lung were determined via ELISA. The data from three independent experiments for each group are expressed as the mean ± SD (n = 5–8 from 3 independent experiments). *p < 0.05.

Close modal

Measurement of CD4+T Cell Expression in BLN Using Flow Cytometry

Based on the observed eosinophil and neutrophil airway infiltration in stress/tolerized mice, we analyzed the T-cell subsets in the BLN lymphocytes on day 20 using flow cytometry (the gating strategy shown in Fig. 3a). As expected, we observed that the Treg cell population in tolerized mice was significantly higher than that in the other groups (shown in Fig. 3b, c). These results suggest that the increased Treg cell level characterizes the occurrence of tolerance. Notably, Treg cell levels in tolerized mice were suppressed by stress exposure. Th17 cells were predominant in the stressed/tolerized group compared to other groups. These results suggest that infiltration of neutrophils in the airways of stress/tolerized mice is induced by inhibiting Treg cells and activating Th17 cells. Based on these results, we considered the possibility that Th17 cells are involved in Treg cell depletion in stress/tolerized mice. To examine this possibility, we used IL-17A knockout (KO) mice. We investigated airway inflammation induced by OVA in stress/tolerized mice. The number of neutrophils was significantly decreased in the IL-17A KO mice. However, the number of eosinophils was not significantly different between the WT and IL-17AKO mice (shown in online suppl. Fig. S1; for all online suppl. material, see www.karger.com/doi/10.1159/000529108). These results suggest that the IL-17A is not directly involved in the reduction of Treg cell development, and conversely, the Treg depletion induced IL-17A production due to Th17 development and neutrophilic airway inflammation. Therefore, we considered that additional upstream immunological changes might have contributed to these observations.

Fig. 3.

Flow cytometry analysis of T-cell subsets in BLN of asthmatic mice. a The flow cytometric gating strategies used to evaluate T cell subsets. b, c The percentages (b) and cell numbers (c) of CD3+CD4+IL-17A+/CD3+CD4+, CD3+CD4+IL4+/CD3+CD4+, and CD3+CD4+FOXP3+/CD3+CD4+ were determined, respectively, in BLN cells. The data from three independent experiments for each group are expressed as the mean ± SD (n = 4–6 from independent experiments). *p < 0.05, and **p < 0.01.

Fig. 3.

Flow cytometry analysis of T-cell subsets in BLN of asthmatic mice. a The flow cytometric gating strategies used to evaluate T cell subsets. b, c The percentages (b) and cell numbers (c) of CD3+CD4+IL-17A+/CD3+CD4+, CD3+CD4+IL4+/CD3+CD4+, and CD3+CD4+FOXP3+/CD3+CD4+ were determined, respectively, in BLN cells. The data from three independent experiments for each group are expressed as the mean ± SD (n = 4–6 from independent experiments). *p < 0.05, and **p < 0.01.

Close modal

Comprehensive Analysis via Microarray and Quantitative Changes in Gene Expression via qPCR

Based on our results, we hypothesized that exposure to stress triggers immune changes. Therefore, we comprehensively investigated the gene expression following stress exposure. The results of comprehensive microarray analysis of BLN of mice from the stressed/tolerized and tolerized groups with immune tolerance on day 6 and day 3 are shown in a heatmap (shown in Fig. 4a). Genes satisfying log ratio >0.9 and log ratio < −0.9 were extracted from the microarray data in the heatmap. From there, we extracted genes that affect eosinophils, neutrophil inflammation, and immune tolerance, and may be involved in this experimental system. Other genes were also altered, but this time, we focused on eosinophilic and neutrophil inflammation. Interestingly, we found that Rorc gene expression in BLN increased, whereas Icos expression decreased during stress exposure. To confirm the reproducibility of these results, we analyzed the difference in gene expression using qPCR, and obtained results similar to those of the microarray (shown in Fig. 4b). The results obtained from the comprehensive microarray and qPCR analysis demonstrated that transcription factors and co-stimulatory receptor were associated with the pathophysiology of Th2- and Th17-dominant asthma.

Fig. 4.

Comprehensive microarray analysis and qPCR analysis in BLN before antigen sensitization. a Heatmap depicting cytokines and chemokines in BLN before antigen sensitization (day −2). b The qPCR assay was employed to test the reproducibility of Rorc gene and Icos gene, which are implicated in eosinophilic inflammation, neutrophilic inflammation, and immunological tolerance and showed characteristic changes from the results obtained by the microarray analysis (n = 4 from independent experiments). *p < 0.05.

Fig. 4.

Comprehensive microarray analysis and qPCR analysis in BLN before antigen sensitization. a Heatmap depicting cytokines and chemokines in BLN before antigen sensitization (day −2). b The qPCR assay was employed to test the reproducibility of Rorc gene and Icos gene, which are implicated in eosinophilic inflammation, neutrophilic inflammation, and immunological tolerance and showed characteristic changes from the results obtained by the microarray analysis (n = 4 from independent experiments). *p < 0.05.

Close modal

Effects of Stress Exposure and OVA Inhalation on IL-1β Expression in Lung

Rorc gene expression is elevated upon stimulation with the pro-inflammatory cytokine IL-1β [19, 20]. Furthermore, IL-1β is a key cytokine that contributes to Th17 cell differentiation [21]. Because stress exposure increased Rorc gene expression in the BLN, we analyzed whether IL-1β is also involved in airway inflammation in stress/tolerized mice. We first compared IL-1β secretion in the lungs immediately after final stress exposure (shown in Fig. 5a). OVA exposure in the tolerized mice group had no effects on the expression of IL-1β, whereas stress exposure increased IL-1β expression in the tolerized mice but not in the non-tolerized mice (shown in Fig. 5b). This result suggests that IL-1β expression need the stress and antigen exposures. Therefore, an increase of IL-1β in stressed/tolerized mice may contribute to Th17 differentiation and stress-induced immune tolerance breakdown.

Fig. 5.

Expression level of IL-1β due to psychological stress before antigen sensitization. IL-1β levels in the lungs were measured via ELISA. Data are expressed as the mean ± SD (n = 3–9 from independent experiments). **p < 0.01.

Fig. 5.

Expression level of IL-1β due to psychological stress before antigen sensitization. IL-1β levels in the lungs were measured via ELISA. Data are expressed as the mean ± SD (n = 3–9 from independent experiments). **p < 0.01.

Close modal

Effects of IL-1 Receptor Antagonist on Stress-Induced Airway Inflammation

In the lung environment, IL-1 receptor (IL-1R) signaling is required for producing IL-17A by mouse CD4+ T cells [21‒23] and facilitates IL-1β in maintaining the Th17 response. To investigate the relationship between IL-1β expression in stress/tolerized mice and the development of neutrophilic and eosinophilic airway inflammation, we administered an IL-1R antagonist (IL-1RA) before each stress exposure. Mice were divided into four groups: asthma, tolerance, stressed/tolerized, and stressed/tolerized/IL-1RA mice. Therefore, we evaluated the effects of IL-1RA on airway inflammation. Interestingly, we observed that neutrophilic and eosinophilic airway inflammation in stress/tolerized mice was abolished upon IL-1RA administration (shown in Fig. 6b). The CD4 population change in the stressed/tolerized group was abolished by IL-1RA administration and returned to that of the tolerized group (shown in Fig. 6c). This result strongly indicates that psychological stress-induced expression of IL-1β suppressed the induction of immune tolerance, resulting in neutrophilic and eosinophilic airway inflammation.

Fig. 6.

Effect of IL-1RA on inflammatory cells during immune tolerance induction. a Schematic representation of protocols used for tolerance, stress exposure, sensitization, and antigen challenge. IL-1RA was administered immediately before daily psychological stress during immune tolerance induction. The cells in each of the four groups of mice were counted. b The respective cells were total inflammatory cells, eosinophils, neutrophils, and lymphocytes. Data from three independent experiments for each group are expressed as mean ± SD (n = 3–6 from independent experiments). *p < 0.05 and **p < 0.01. c The percentages of CD3+CD4+IL-17A+/CD3+CD4+, CD3+CD4+IL4+/CD3+CD4+, and CD3+CD4+FOXP3+/CD3+CD4+ were determined. The flow cytometric gating strategies are shown in Figure 2 b. The data from three independent experiments for each group are expressed as the mean ± SD (n = 5–6 from independent experiments). *p < 0.05 and **p < 0.01.

Fig. 6.

Effect of IL-1RA on inflammatory cells during immune tolerance induction. a Schematic representation of protocols used for tolerance, stress exposure, sensitization, and antigen challenge. IL-1RA was administered immediately before daily psychological stress during immune tolerance induction. The cells in each of the four groups of mice were counted. b The respective cells were total inflammatory cells, eosinophils, neutrophils, and lymphocytes. Data from three independent experiments for each group are expressed as mean ± SD (n = 3–6 from independent experiments). *p < 0.05 and **p < 0.01. c The percentages of CD3+CD4+IL-17A+/CD3+CD4+, CD3+CD4+IL4+/CD3+CD4+, and CD3+CD4+FOXP3+/CD3+CD4+ were determined. The flow cytometric gating strategies are shown in Figure 2 b. The data from three independent experiments for each group are expressed as the mean ± SD (n = 5–6 from independent experiments). *p < 0.05 and **p < 0.01.

Close modal

In this study, we discovered that persistent psychological stress hinders immunological tolerance induction and triggers the development of Th17- and Th2-dominant asthma. Our previous studies have shown that stress impairs the establishment of immune tolerance and causes asthma in mouse models. The expression of eosinophils and neutrophils in each group revealed increased eosinophils in the asthma mice group, indicating a Th2-dominant airway inflammation. Similarly, stressed mice also developed Th2-dominant asthma. In tolerized mice, eosinophils were significantly reduced compared to that in asthmatic mice. This indicates that immune tolerance suppresses the onset of Th2-dominant asthma. However, both eosinophils and neutrophils increased in stressed/tolerized mice compared to tolerized mice. This suggests the suppression of immune tolerance and promotion of the onset of Th2-dominant asthma and neutrophilic asthma, in which Th17 cells are increased. There was no difference in both eosinophil and neutrophil levels between asthmatic and stressed mice. This suggests that stress does not affect Th2-dominant asthma. However, when comparing tolerized and stressed/tolerized mice, eosinophils and neutrophils were predominantly increased in stressed/tolerized mice. This indicates the development of Th2-dominant asthma in addition to Th17-dominant asthma, which further implies that stress does not induce immune tolerance but causes the development of neutrophilic asthma.

To confirm these results, T-cell subsets in BLN were confirmed in post-sensitized mice. The confirmed subsets were Th2 (IL-4), Treg (FOXP3), and Th17 (IL-17A) cells. The results showed that in tolerized mice, Treg cells were the highest, Th2 cells were significantly low, and Th17 cells were similar compared to the non-tolerized mice. Therefore, eosinophil inflammatory reaction is suppressed by immune tolerance. Stress/tolerized mice exhibited decreased Treg cell and increased Th2 and Th17 cell count. This indicates that stress suppresses immune tolerance and exacerbates inflammatory responses in neutrophils and eosinophils.

These results were obtained by comprehensively performing gene analysis using a microarray (shown in Fig. 4). Based on the results of the analysis, the following can be considered: ICOS is involved in the production, proliferation, and survival of Treg cells [24]. Stress exposure significantly decreased Icos expression. These results indicate that stress suppresses Treg cell differentiation, resulting in an environment where immune tolerance is less likely to occur. The mechanism underlying Th17 lymphocyte differentiation involves the binding of IL-6 to signaling and transcriptional activator 3 (STAT3), which directly binds to the chromatin of Th17 cells and activates RORγt [25, 26]. The expression of Rorc gene significantly increases, indicating an increase in the Th17 cell level. Since Th17 cells characterize a neutrophilic inflammatory response, stress is considered to cause neutrophilic asthma [26] by eliciting a neutrophilic inflammatory response. The fact that Ccl24 and Rorc expression is increased by stress suggests neutrophilic and eosinophilic inflammatory reactions.

IL-1β is involved in early Th17 cell differentiation [27]. As described above, the differentiation of dendritic cells into Th17 cells makes it difficult for immune tolerance to occur and creates an environment where eosinophil and neutrophil inflammation are likely to occur. In this study, IL-1β was produced only when stress and OVA exposure were combined, but not in cases of stress exposure alone or OVA exposure alone (Fig. 5). The results suggest that the increased IL-1β expression in lung may be caused by a synergistic effect between brain cell activity in response to stress exposure and peripheral cell activity in response to antigen exposure, as psychological stress has been reported to activate microglia in the brain and increase IL-1β production [28]. In addition, antigen stimulation has also been reported to induce IL-1β production from lung tissue macrophages and airway epithelial cells [29]. Therefore, we investigated the effects of stress by administering IL-1RA. Using IL-1β as an activator increases the activity of the transcription factor RORγt [26]. Therefore, the action of IL-1 could be canceled by administering IL-1RA, which has an antagonistic action. We found that the number of inflammatory cells and the development of Th17 decreased in stressed/tolerized/IL-1RA mice. This suggests that stress-induced action was inhibited by IL-1RA, which suppresses Th17 cell differentiation.

This current study found that stress before sensitization increases cytokines involved in eosinophil and neutrophil inflammation, thereby worsening asthma symptoms. Furthermore, the administration of IL-1RA reduces inflammatory substances that exacerbate eosinophilic and neutrophilic asthma symptoms. Currently, research is being conducted in animal models; however, further clinical trials involving humans are warranted to determine the precise effects of these cytokines in the pathophysiology of human asthma phenotypes.

All experimental procedures involving animals were approved by the Committee of Animal Experiments at Tohoku Medical and Pharmaceutical University (approval numbers: 16002-cn, 17004-cn, A2040, A2207). All experiments were performed in compliance with relevant institutional guidelines.

The authors declare that they have no conflict of interest.

This study was supported by a Japan Society for the Promotion of Science (JSPS) Grant-in-Aid for Young Scientists (B) (No. 17K16212). The funders played no role in the study design, collection, analysis, interpretation of the data, or preparation of the manuscript.

S.S., T.K., and T.T. planned the experiments, wrote the manuscript, and performed the majority of the experiments. S.S. and E.I. conducted the qPCR and flow cytometric analysis. J.S. conducted the ELISA assay. K.T. and T.M. induced asthma models and conducted tissue processing and BAL. Y.M., F.I., and M.T. discussed the experiments with T.K.

All data generated or analyzed during this study are included in this article and its supplementary material files. Further inquiries may be directed to the corresponding author.

1.
Mattiuzzi
C
,
Lippi
G
.
Worldwide asthma epidemiology: insights from the global health data exchange database
.
Int Forum Allergy Rhinol
.
2020 Jan
10
1
75
80
.
2.
Wenzel
SE
.
Asthma phenotypes: the evolution from clinical to molecular approaches
.
Nat Med
.
2012 May 4
18
5
716
25
.
3.
Yonas
MA
,
Lange
NE
,
Celedon
JC
.
Psychosocial stress and asthma morbidity
.
Curr Opin Allergy Clin Immunol
.
2012 Apr
12
2
202
10
.
4.
Fahy
JV
.
Type 2 inflammation in asthma--present in most, absent in many
.
Nat Rev Immunol
.
2015 Jan
15
1
57
65
.
5.
Kawano
T
,
Ouchi
R
,
Ishigaki
T
,
Masuda
C
,
Miyasaka
T
,
Ohkawara
Y
.
Increased susceptibility to allergic asthma with the impairment of respiratory tolerance caused by psychological stress
.
Int Arch Allergy Immunol
.
2018
;
177
(
1
):
1
15
.
6.
Akbari
O
,
DeKruyff
RH
,
Umetsu
DT
.
Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen
.
Nat Immunol
.
2001 Aug
2
8
725
31
.
7.
Kim
DI
,
Song
MK
,
Lee
K
.
Comparison of asthma phenotypes in OVA-induced mice challenged via inhaled and intranasal routes
.
BMC Pulm Med
.
2019 Dec 10
19
1
241
.
8.
Sakaguchi
S
,
Yamaguchi
T
,
Nomura
T
,
Ono
M
.
Regulatory T cells and immune tolerance
.
Cell
.
2008 May 30
133
5
775
87
.
9.
Ouchi
R
,
Kawano
T
,
Yoshida
H
,
Ishii
M
,
Miyasaka
T
,
Ohkawara
Y
.
Maternal separation as early-life stress causes enhanced allergic airway responses by inhibiting respiratory tolerance in mice
.
Tohoku J Exp Med
.
2018
;
246
(
3
):
155
65
.
10.
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 May
40
5
1232
40
.
11.
Sakaguchi
S
,
Miyara
M
,
Costantino
CM
,
Hafler
DA
.
FOXP3+ regulatory T cells in the human immune system
.
Nat Rev Immunol
.
2010
;
10
(
7
):
490
500
.
12.
Yasuda
K
,
Takeuchi
Y
,
Hirota
K
.
The pathogenicity of Th17 cells in autoimmune diseases
.
Semin Immunopathol
.
2019 May
41
3
283
97
.
13.
Minns
D
,
Smith
KJ
,
Alessandrini
V
,
Hardisty
G
,
Melrose
L
,
Jackson-Jones
L
.
The neutrophil antimicrobial peptide cathelicidin promotes Th17 differentiation
.
Nat Commun
.
2021 Feb 24
12
1
1285
.
14.
Ackerman
KD
,
Martino
M
,
Heyman
R
,
Moyna
NM
,
Rabin
BS
.
Stressor-induced alteration of cytokine production in multiple sclerosis patients and controls
.
Psychosom Med
.
1998 Jul–Aug
60
4
484
91
.
15.
Shao
BZ
,
Xu
ZQ
,
Han
BZ
,
Su
DF
,
Liu
C
.
NLRP3 inflammasome and its inhibitors: a review
.
Front Pharmacol
.
2015
;
6
:
262
.
16.
Wang
J
,
Charboneau
R
,
Barke
RA
,
Loh
HH
,
Roy
S
.
Mu-opioid receptor mediates chronic restraint stress-induced lymphocyte apoptosis
.
J Immunol
.
2002 Oct 1
169
7
3630
6
.
17.
Okuyama
K
,
Dobashi
K
,
Miyasaka
T
,
Yamazaki
N
,
Kikuchi
T
,
Sora
I
.
The involvement of glucocorticoids in psychological stress-induced exacerbations of experimental allergic asthma
.
Int Arch Allergy Immunol
.
2014
;
163
(
4
):
297
306
.
18.
Ito
C
,
Okuyama-Dobashi
K
,
Miyasaka
T
,
Masuda
C
,
Sato
M
,
Kawano
T
.
CD8+ T cells mediate female-dominant IL-4 production and airway inflammation in allergic asthma
.
PLoS One
.
2015
;
10
(
10
):
e0140808
.
19.
Zielinski
CE
,
Mele
F
,
Aschenbrenner
D
,
Jarrossay
D
,
Ronchi
F
,
Gattorno
M
.
Pathogen-induced human TH17 cells produce IFN-γ or IL-10 and are regulated by IL-1β
.
Nature
.
2012 Apr 26
484
7395
514
8
.
20.
Uchiyama
R
,
Yonehara
S
,
Taniguchi
S
,
Ishido
S
,
Ishii
KJ
,
Tsutsui
H
.
Inflammasome and fas-mediated IL-1β contributes to Th17/Th1 cell induction in pathogenic bacterial infection in vivo
.
J Immunol
.
2017 Aug 1
199
3
1122
30
.
21.
Duhen
T
,
Campbell
DJ
.
IL-1β promotes the differentiation of polyfunctional human CCR6+CXCR3+ Th1/17 cells that are specific for pathogenic and commensal microbes
.
J Immunol
.
2014 Jul 1
193
1
120
9
.
22.
Reed
M
,
Morris
SH
,
Owczarczyk
AB
,
Lukacs
NW
.
Deficiency of autophagy protein Map1-LC3b mediates IL-17-dependent lung pathology during respiratory viral infection via ER stress-associated IL-1
.
Mucosal Immunol
.
2015 Sep
8
5
1118
30
.
23.
Jain
R
,
Chen
Y
,
Kanno
Y
,
Joyce-Shaikh
B
,
Vahedi
G
,
Hirahara
K
.
Interleukin-23-Induced transcription factor blimp-1 promotes pathogenicity of T helper 17 cells
.
Immunity
.
2016 Jan 19
44
1
131
42
.
24.
Li
DY
,
Xiong
XZ
.
ICOS(+) tregs: a functional subset of tregs in immune diseases
.
Front Immunol
.
2020
;
11
:
2104
.
25.
Ivanov
II
,
McKenzie
BS
,
Zhou
L
,
Tadokoro
CE
,
Lepelley
A
,
Lafaille
JJ
.
The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells
.
Cell
.
2006 Sep 22
126
6
1121
33
.
26.
Leija-Martínez
JJ
,
Huang
F
,
Del-Río-Navarro
BE
,
Sanchéz-Muñoz
F
,
Romero-Nava
R
,
Muñoz-Hernandez
O
.
Decreased methylation profiles in the TNFA gene promoters in type 1 macrophages and in the IL17A and RORC gene promoters in Th17 lymphocytes have a causal association with non-atopic asthma caused by obesity: a hypothesis
.
Med Hypotheses
.
2020 Jan
134
109527
.
27.
Ikeda
S
,
Saijo
S
,
Murayama
MA
,
Shimizu
K
,
Akitsu
A
,
Iwakura
Y
.
Excess IL-1 signaling enhances the development of Th17 cells by downregulating TGF-β-induced Foxp3 expression
.
J Immunol
.
2014 Feb 15
192
4
1449
58
.
28.
McKim
DB
,
Weber
MD
,
Niraula
A
,
Sawicki
CM
,
Liu
X
,
Jarrett
BL
.
Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety
.
Mol Psychiatry
.
2018 Jun
23
6
1421
31
.
29.
Mattiuzzi
C
,
Lippi
G
,
Couillin
I
,
Tan
Z
,
Zheng
SG
,
Erard
F
.
Worldwide asthma epidemiology: insights from the global health data exchange databaseMattiuzzi C, lippi G. Worldwide asthma epidemiology: insights from the global health data exchange database
.
J Mol Cell BiolInt Forum Allergy Rhinol
.
2012 Feb 2020 Jan
410
11
375
1080
.

Additional information

Edited by: H.-U. Simon, Bern.