2023 Impact Factor
Asthma is a chronic inflammatory lung disease characterized by airway hyperresponsiveness, inflammation, and respiratory remodeling, often observed in pediatric and adult patients, causing significant morbidity, mortality, and high economic burden worldwide (Gans and Gavrilova, 2020). There are several types of asthma, including allergic, seasonal, non-allergic, occupational, exercise-induced, difficult-to-control, and severe, each with its own set of triggers (Padem and Saltoun, 2019).
Allergic asthma is the most common form of asthma and is present in 50-80% of all asthma cases. Common triggers of allergic asthma are mostly attributed to interactions between environmental exposure and genetic susceptibility. These include environmental factors (dust mites, animal hair, pollen, mold, obesity, stress, and other allergic sensitizations) and genetic factors (asthma susceptibility loci) (Toskala and Kennedy, 2015). When patients with asthma are exposed to allergens, the rapid release of pro-inflammatory mediators causes contraction of airway smooth muscle, swelling of the mucosal lining, and oversecretion of mucus, thereby blocking the narrowed airway and inducing difficulty in breathing up to extreme levels (Fixman
The pathophysiology of asthma involves a complex interplay between the innate and adaptive immune systems, thereby stimulating chronic airway inflammation (Kim
Neurotrophins mediate the differentiation, growth, and survival of responsive neurons by binding to two types of cell surface receptors: Trk tyrosine kinase receptors and p75 neurotrophin receptor (p75NTR) (Kaplan and Miller, 2000). Nerve growth factor (NGF) is a member of the neurotrophin family that induces bronchial hyperresponsiveness by increasing sensory innervation. These changes are expected to induce the migration and activation of inflammatory cells (Quarcoo
Asthma is a lifelong condition that can be managed with medication and avoidance of triggers that initiate asthma attacks. Glucocorticoid anti-inflammatory drugs are currently recommended in clinical practice as anti-asthma drugs for patients with mild-to-moderate asthma; however, the daily inhalation of glucocorticoids can lead to poor patient compliance and life-threatening events. Moreover, the long-term use of steroidal anti-inflammatory drugs can cause serious complications such as pneumonia, fractures, hyperglycemia, and cataracts (Li
Natural products have been widely used in the treatment and prevention of human diseases for thousands of years owing to their effectiveness, wide margins of safety, and multi-targeting properties. Previously, our group investigated the composition and efficacy of a traditional herbal medicine called ‘So Cheong Ryong Tang’ which is the water extract of various herbs including
With our recent successful application of BS012 in an atopic dermatitis experimental model (Lee
BS012 comprises a combination of three herbal medicines: PG, CC, and AS. The three herbal medicines were purchased from Kyungdong Market (Woori Herb), Seoul, Korea. The materials were verified and prepared as BS012 by our research collaborator Prof. Hocheol Kim at the Department of Traditional Medicine, Kyung Hee University (Seoul, Korea). The BS012 extract was prepared as described in our previous report (Lee
In general, female mice have more pronounced B cell-mediated immunity than age-matched males, and therefore have higher Ig levels and stronger antibody responses to various foreign antigens. Various studies have suggested that female mice are more susceptible to allergic airway inflammation than male mice (Melgert
After one week of acclimatization, the mice were randomly divided into five experimental groups (n=8 per group) as follows: control, OVA, OVA+BS012 (100 and 200 mg/kg), and OVA+dexamethasone (1.5 mg/kg). Except for the control group, all other mice were sensitized with an intraperitoneal injection of OVA (1 mg/kg) along with aluminum hydroxide (200 mg in 1 mL of saline) on days 1 and 8. Dexamethasone (1.5 mg/kg body weight) was intraperitoneally administered as a positive control from day 15, once daily, for 4 days. BS012 was orally administered at doses of 100 and 200 mg/kg body weight from day 1, once daily for 19 days. All animals, except for the control group, were challenged with 5% (w/v) OVA solution aerosolized using an ultrasonic nebulizer from day 15 onwards for 30 min per day for 4 days. On day 19, all animals were anesthetized and whole blood was collected from the abdominal aorta to separate the plasma for biochemical analysis. The lungs were lavaged three times with cold saline through a tracheal cannula to collect bronchoalveolar lavage fluid (BALF). BALF was further centrifuged to collect the supernatant, which was stored at –80°C until further analysis. Part of the lung tissue was stored in 10% formalin at room temperature for histopathological analysis and the rest was snap frozen and kept at –80°C until protein extraction. The experimental schedule is shown in Fig. 1A.
Levels of IL-4 (R&D Systems, MN, USA, Cat. M4000B) and IL-5 (cat. M5000) in BALF, and OVA-specific IgE (BioLegend, San Diego, CA, USA, Cat. 439807) in the plasma were measured using an enzyme-linked immunosorbent assay (ELISA) following the manufacturer’s instructions. Specific monoclonal antibodies were pre-coated onto the microplates. Standards and samples were pipetted into the wells and any specific antigen present was bound by the immobilized antibody. After washing away unbound substances, a specific enzyme-linked monoclonal antibody was added to the wells. Following washing to remove any unbound antibody-enzyme reagent, a colorimetric substrate was added to the wells to form a colored solution when catalyzed by the enzyme. The absorbance was measured at 450 nm using a microplate reader.
Lung samples fixed in 10% formalin were embedded in paraffin and sliced into 4 μm thick sections. Lung histology was further confirmed using hematoxylin and eosin (H&E) staining and the thickness of the bronchial epithelium was measured using ImageJ software. Additionally, to identify the degree of goblet cell hyperplasia and mucus production in the airway, Periodic Acid Schiff (PAS) staining was performed, and the number of PAS-positive cells was counted using ImageJ software (Figi, Paris, France).
Total protein was extracted from lung tissue using PRO-PREP™ protein extraction solution (iNtRON Biotechnology, Seongnam, Korea) by centrifuging the lysate at 13,000 rpm for 15 min at 4°C to collect the protein-containing supernatant in a new tube. The protein concentration was quantified using a bovine serum albumin (BSA) kit (Cat. #5000207) following the manufacturer’s instructions. Protein samples were separated using sodium dodecyl sulfate polyacrylamide gel electrophoresis gel and then transferred to 0.2 μM poly-vinylidene difluoride membrane. After blocking with Every Blot Blocking Buffer (Bio-Rad, CA, USA, Cat. #12010020), the membranes were incubated with primary antibodies overnight at 4°C and incubated with appropriate secondary antibodies for 1 h at room temperature. The signal intensity was imaged and quantified using a LAS-4000 system (Fuji Film, Tokyo, Japan). The primary antibodies (NGF (#52918, Abcam), p75NTR (#4201), P-JNK (#9251), JNK (#9252), P-c-jun (#3270), c-jun (#9165), Bcl-xl (#2764), P-Bad (#5284), Bad (#9292), Bax (#2772), Cytochrome c (#11940), cleaved Caspase-9 (#9509), Caspase-9 (#9504), cleaved Caspase-3 (#9664), Caspase-3 (#9662), and GAPDH (#2118)) and secondary antibodies (rabbit IgG (#7074)) were purchased from Cell Signaling Technology (MA, USA).
Bone marrow cells were obtained from the femurs and tibias of naïve mice and cultured in complete RPMI medium supplemented with 2.5% HEPES and 20 ng/mL GM-CSF to induce differentiation. Flow cytometry confirmed that more than 90% of the cells were CD11c+ dendritic cells (DC). Dendritic cells (DC) were prepared for co-culture by CD11c MACS enrichment of bone marrow-derived dendritic cells. CD4+ T cells were enriched by collecting and pooling spleens and peripheral lymph nodes from 6-week male C57BL/6N mice, followed by CD4 MACS enrichment (Miltenyi Biotec, Bergisch, Germany). The enriched samples were identified and flow-sorted for naïve CD4+-gated CD4+CD44lowCD25-CD62Lhi cells using a FACSAria III cell sorter. For DC - CD4+ T cells co-culture assays, cells were co-cultured in complete media with soluble anti-CD3ε Ab (clone 145-2C11), anti-IFNγ (clone XMG1.2), IL-2, IL-4 in the presence of N-isobutyl-dodecatetraenamide for 3 days. For intracellular cytokine analysis, the cell suspension was incubated with PMA (Cat# P1585, Sigma-Aldrich, St. Louis, MO, USA), ionomycin (Cat# I9657, Sigma-Aldrich), GolgiStop (Cat#00450551, Thermo Fisher, MA, USA), and GolgiPlug (Cat# 00450651, Thermo Fisher) for 3 h. Fixation and permeabilization were performed using an Intracellular Fixation & Permeabilization Buffer set (Cat# 88-8824-00, Thermo Fisher) following the manufacturer’s protocol. The samples were analyzed using a FACSLyric (BD Biosciences, CA, USA). The antibodies used for flow cytometry analysis are listed in Supplementary Table 1.
The data were presented as the mean ± standard error of the mean (SEM). Statistical analyses were performed using the Prism 10 software (GraphPad Software, Inc., San Diego, CA, USA). After the normal distribution test, differences among the groups were analyzed using one-way analysis of variance, followed by Tukey’s multiple comparison post-hoc test. Statistical significance was set at 0.05.
The plasma metabolome was extracted according to the protocol outlined by Lee (Lee
Prepared samples were analyzed using Orbitrap Exploris™ 120 Mass spectrometer coupled with Vanquish Flex UHPLC system (Thermo Fisher Scientific, San Jose, CA, USA) at the Biopolymer Research Center for Advanced Materials (BRCAM). Samples were injected into a randomized analytical batch along with quality control (QC) samples. The QC samples consisted of equal volumes of samples for conditioning and were included in every 10 samples throughout the batch. The instrumental parameters of UPLC-Orbitrap-MS have been described previously (Lee
To investigate the effects of BS012 on OVA-mediated allergic responses in mice, plasma OVA-specific IgE levels were measured. As Fig. 1B shows, secretion of OVA-specific IgE in the plasma was significantly increased in the OVA-induced group (21.8 ± 1.6 ng/mL) compared with the control group (0.1 ± 0.0 ng/mL). The mice treated with BS012 100 and 200 mg/kg showed significantly down-regulated levels of OVA-specific IgE (14.6 ± 0.9 ng/mL and 13.8 ± 1.0 ng/mL, respectively) in the plasma compared with the OVA group mice.
Among the various Th2-specific cytokines secreted, the levels of IL-4 (Fig. 1C) and IL-5 (Fig. 1D) were measured in BALF samples. The level of IL-4 was significantly increased in the OVA-induced mice (188.2 ± 31.9 ng/mL) compared with the control group (4.4 ± 0.4 ng/mL). The mice treated with BS012 100 (123.5 ± 21.0 ng/mL) and 200 (88.0 ± 14.1 ng/mL) mg/kg showed a significant reduction in IL-4 level compared with the OVA group mice in a dose-dependent manner. Similar to the IL-4 results, IL-5 level in BALF was also increased significantly in the OVA group (313.1 ± 31.3 ng/mL) compared with the control group (10.2 ± 1.2 ng/mL). IL-5 level was markedly decreased in the mice treated with BS012 at doses of 100 (86.4 ± 14.5 ng/mL) and 200 (45.7 ± 10.9 ng/mL) mg/kg compared to the control group. As expected, dexamethasone injection, which was used as a positive control, significantly suppressed the levels of OVA-specific IgE, IL-4, and IL-5 by 65.6%, 81.0%, and 84.0%, respectively, compared to the OVA group.
Structural changes were observed by the histological evaluation of H&E-stained lung tissue sections (Fig. 2A). The mice in the control group had smooth and intact bronchial structures. The thickness of the bronchus was normal (11.24 ± 1.5 μm) without inflammatory infiltration around the bronchioles. Compared with the control group, there was a significant increase in the infiltration of inflammatory cells around the bronchioles in the OVA-induced group. Such change was accompanied by prominent thickening (three times) of the bronchiole walls in the OVA-induced group (35.9 ± 5.3 μm) compared to the control group (Fig. 2B). BS012 treatment significantly reduced these histological changes in a dose-dependent manner. Reduced inflammatory cell infiltration was visible and reduced thickening of the bronchial epithelium (16.0 ± 2.6 μm) was statistically significant in the BS012 200 mg/kg treatment group. Such a reduction in the epithelium was also observed in the dexamethasone-treated group.
OVA-induced goblet cell hyperplasia and mucus production in lung tissue were examined by PAS staining. As shown in Fig. 2C, the control group exhibited a smooth mucosal layer at the bronchioles with no signs of mucus formation. However, with the OVA challenge, mucus hypersecretion was observed, with a marked increase in goblet cell hyperplasia in the bronchioles. In addition, as shown in Fig. 2D, significantly increased PAS-positive cells were counted in the OVA-induced group, and approximately 45 PAS-positive cells/mm2 were detected compared to the control group (3 PAS-positive cells/mm2). Dexamethasone and BS012 200 mg/kg treatments both showed a substantial reduction in PAS-positive cell counts (5 and 4 PAS-positive cells/mm2, respectively) to the normal level along with decreased mucus secretion and goblet cell hyperplasia in the bronchioles.
To determine the effect of BS012 on the apoptotic pathway in OVA-induced asthmatic mice, proteins related to the NGF-mediated JNK signaling pathway were measured in the lung tissue using western blotting (Fig. 3). Challenge with OVA increased the protein level of NGF up to 184.8 ± 5.8% in the OVA group compared with the control group (Fig. 3A). BS012 treatment at 200 mg/kg significantly decreased the NGF protein expression level to 134.4 ± 4.7% when compared with the control group. Dexamethasone injection showed a similar effect as the BS012 200 mg/kg treatment reaching 132.0 ± 6.6% compared to the control group. However, a lower decrease was observed in the BS012 100 mg/kg treated group, at 161.1 ± 18.7% of the control group. Meanwhile, the protein expression level of p75NTR was also significantly upregulated with OVA challenge when compared to the control group exhibiting 392.2 ± 27.4% of the control group (Fig. 3B). Dexamethasone and BS012 200 mg/kg treatment significantly reduced this expression level down to 224.0 ± 15.6% and 230.0 ± 16.1% of the control group, respectively. The p75NTR protein expression level also showed a reducing trend in the BS012 100 mg/kg treatment group (324.0 ± 12.0% of the control group); however, this was not statistically significant. Under the challenge of OVA, the protein expression level of JNK was also significantly enhanced, up to 305.4 ± 32.8% of the control group (Fig. 3C). JNK protein expression level was significantly suppressed down with dexamethasone and BS012 200 mg/kg treatment, down to 133.3 ± 8.1% and 169.3 ± 21.1% of the control group, respectively. Administration of 100 mg/kg BS012 resulted in a slight decrease in JNK levels, with no statistical significance, compared to the OVA group. For the c-Jun protein expression as shown in Fig. 3D, exposure to OVA up-regulated its expression level to 169.6 ± 7.0% of the control group. BS012 treatment diminished the c-Jun protein expression in a dose-dependent manner, 100 and 200 mg/kg BS012 both significantly reduced its expression level down to 103.8 ± 5.6% and 55.3 ± 3.5% of the control group, respectively. Injection with dexamethasone also significantly suppressed c-Jun expression down to 85.8 ± 13.6% that of the control group.
We measured the expression of several proteins belonging to the Bcl-2 family (Fig. 4A-4C). We found that protein expression of P-Bad was highly upregulated by the OVA challenge, up to 307.0 ± 6.3% compared with the control group (Fig. 4A). Dexamethasone injection significantly reduced its expression level down to 158.5 ± 18.1% compared to the control group. Administration with BS012 100 and 200 mg/kg suppressed the Bad protein expression in a dose-dependent manner, but only the BS012 200 mg/kg treated group showed statistical significance against the OVA-induced group, at 259.8 ± 9.4% of the control group. Exposure to OVA also highly increased the expression level of Bax protein to 166.5 ± 7.4% of the control group (Fig. 4B). This up-regulation was significantly suppressed in the group injected with dexamethasone, at 95.2 ± 9.1% of the control group. BS012 100 and 200 mg/kg treatment suppressed the expression of Bax down to 130.8 ± 5.5% and 123.9 ± 5.5% of the control group, respectively, statistical significance against the OVA-induced group was found both in these two groups. As Fig. 4C shows, anti-apoptotic protein Bcl-xl was decreased by the OVA challenged, down to 52.8 ± 2.4% of the control group, while dexamethasone injection significantly enhanced its expression level up to 135.0 ± 12.9% of the control group. No apparent difference was observed in the BS012 100 mg/kg treated group when compared to the OVA group. In contrast, administration with BS012 200 mg/kg showed statistical significance when compared with the OVA group, at 97.5 ± 7.2% of the control group.
Next, we analyzed the protein expression levels of cytochrome c, caspase-9, and caspase-3 to further confirm the protective effect of BS012 on the cytochrome c-initiated caspase activation pathway following OVA (Fig. 4D-4F). Significant upregulation of cytochrome-c protein expression was observed in OVA-induced group (491.5 ± 13.8%) compared to the control group (Fig. 4D). High expression level of cytochrome-c was prevented by dexamethasone treatment with statistical significance compared to the OVA group, exhibited 148.3 ± 12.4% of the control group. Even though the cytochrome-c level was not much altered by BS012 100 mg/kg treatment (441.4 ± 17.0%) when compared to the OVA group, BS012 200 mg/kg treatment significantly decreased its level down to 341.4 ± 29.5% of the control group, which was significantly different to the OVA group. For the caspase-9 protein expression as shown in Fig. 4E, exposure to the OVA increased its expression level up to 199.6 ± 10.3% of the control group. Treatment of dexamethasone and BS012 100 mg/kg slightly decreased the caspase-9 level without statistical significance when compared to the OVA group (162.1 ± 5.6% and 185.8 ± 14.4% compared to the control group, respectively). However, a significant decrease in caspase-9 level was found in the BS012 200 mg/kg treated group, down to 130.1 ± 11.8% of the control group. The expression level of caspase-3 protein (Fig. 4F) was significantly increased in the OVA group, up to 1953.0 ± 104.3% of the control group. BS012 200 mg/kg significantly reduced the caspase-3 protein expression down to 975.8 ± 158.7% compared to the control group and it showed statistical significance against the OVA-induced group. Dexamethasone and BS012 100 mg/kg treated groups showed less reduction level of caspase-3 protein expression, at 1188.0 ± 290.8% and 1238.3 ± 138.6% of the control group, respectively.
The plasma metabolite profiles of the OVA-induced asthmatic mice were characterized using UPLC-Orbitrap-MS in both positive and negative ionization scan modes. Multivariate statistical analysis was used to visualize the cross-group comparison of the metabolic patterns of OVA-treated mice following BS012 administration (Fig. 5A, 5B). Clustering of metabolic patterns was not only visible between the control group and OVA-induced asthmatic mice but also among groups treated with dexamethasone and BS012 at different doses, suggesting that they have distinct metabolic profiles. Univariate analysis was used to identify metabolites that showed significant differences between the OVA-induced and BS012-treated groups. As a result, 20 metabolites were identified and most of them belonged to lipid and amino acid metabolism (Fig. 5C). After BS012 administration, the metabolic changes between the OVA-induced and control groups showed an inverse relationship (Supplementary Table 2). Graphs of each metabolite change in plasma are presented in Supplementary Fig. 1.
The lung tissue metabolite profiles of the OVA-induced asthmatic mice were analyzed using the same method (Fig. 6A, 6B). In the lung tissue, the metabolic profile of the OVA-induced asthma group showed significant changes compared with that of the control group. The groups administered dexamethasone and 100 mg/kg BS012 showed similar metabolic profiles, whereas the group administered 200 mg/kg BS012 was clustered. Upon identifying the metabolites contributing to these changes, a total of 26 metabolites were found to be significantly altered in the OVA-induced group compared to the control group and BS012 treated group compared to the OVA-induced asthma group (Supplementary Table 3). The identified metabolites were classified into four classes based on their related metabolism, with the majority categorized as lipid metabolites (Fig. 6C). Most metabolites showed an increasing trend after asthma induction, followed by a decrease after BS012 or Dexamethasone administration, whereas TG levels showed the opposite trend. Graphs of changes in each metabolite in the lung tissue are shown in Supplementary Fig. 2. The overall pathways of metabolic changes induced by OVA and BS012 in the plasma and lung tissues are summarized in Fig. 7.
Among the main components of BS012,
As a complex and chronic inflammatory disorder, several key cellular and molecular mediators are involved in the airway inflammatory response during the progression of allergic asthma. Th2 cells contribute to the immunopathology of allergic asthma by producing various inflammatory mediators, including IL-4 and IL-5, which further contribute to the hallmark features of asthma, such as IgE production, airway inflammation, and remodeling (Lambrecht
Airway remodeling is a pathological feature of chronic asthma that contributes to the clinical manifestations of the disease, such as persistent airflow obstruction, which can be attributed to goblet cell hyperplasia, airway smooth muscle cell proliferation, and fibroblast activation with increased bronchial epithelium thickness (Hough
To explore the specific protective mechanism of BS012 in allergic asthma, we investigated its role BS012 in OVA-induced lung cell apoptosis. Previous studies have shown that NGF tends to increase at the site of inflammation in inflammatory and autoimmune diseases, especially those characterized by the abnormal activation of immune cells and increased cytokine production (Minnone
In plasma metabolomics, which reflects systemic changes in the whole body, amino acid metabolism related to inflammation and energy metabolism was mainly altered after BS012 administration. Phenylalanine metabolism increased after OVA induction and decreased after BS012 administration. This disruption in phenylalanine metabolism is noted not only as a hallmark of atopic asthma exacerbations in children but is also significantly correlated with lung damage in patients with acute respiratory distress syndrome (Cottrill
Another significant metabolic change observed in plasma is related to lipid metabolism. Decreased sphingolipid metabolism is associated with an increased risk of developing asthma in children and lower concentrations of key phosphosphingolipids, such as sphingosine-1-phosphate, are associated with increased airway resistance (Rago
We investigated the effect of BS012 on lung tissue metabolism, which is directly influenced by asthma symptoms, and observed clear changes in inflammation-related lipid metabolism. Notably, the upregulation of glycerophospholipids was observed after OVA induction. Several studies have reported the involvement of LPCs in acute lung injury and chronic inflammatory lung diseases such as asthma. Elevation of LPC in the lungs of asthmatic subjects and the promotion of inflammatory responses by exogenous LPC administration in the lungs of animal models have shown a correlation between LPC and inflammatory damage in the lungs (Nishiyama
l-Carnitine plays a crucial role in enabling the transport of fatty acyl-CoA into the mitochondria for fatty acid oxidation. Inhibition of this process results in the accumulation of acylcarnitine derivatives (Al-Biltagi
In addition to changes in lipid metabolism, alterations in purine metabolism have also been observed in the lung tissue. The observed increase in inosine levels in bronchial asthma models can occur when adenosine is metabolized to inosine, potentially because of the breakdown of ATP induced during inflammation in allergic asthma (Moon
We performed additional experiments to determine which of the major components of BS012, identified in our previous study, influenced this mechanism of action (Lee
In conclusion, this study demonstrated that BS012, an herbal remedy comprising
This research was funded by the Bio-Synergy Research Project [NRF2013M3A9C4078145] [NRF 2012M3A9C4048794] of the Ministry of Science, ICT, and Future Planning through the National Research Foundation of the Korea Institute of Science and Technology Institutional Program [2E31623].
The authors declare that they have no competing interests.