Open Access

SIRT3 regulates bronchial epithelium apoptosis and aggravates airway inflammation in asthma

  • Authors:
    • Jie Song
    • Jinxiang Wang
  • View Affiliations

  • Published online on: February 28, 2022     https://doi.org/10.3892/mmr.2022.12660
  • Article Number: 144
  • Copyright: © Song et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sirtuin (SIRT)3 is closely related to inflammation and apoptosis and studies have described this relationship, including in the lungs. However, the expression of SIRT3 and its effect on apoptosis and inflammation in bronchial tissue in asthma remains to be elucidated. The present study found that SIRT3 expression decreased in the bronchial tissues of asthmatic mice and its upregulation could not only reduce increased bronchial epithelial cells apoptosis in the asthmatic mice but also significantly decreased the elevated expression of cytokines (TNF‑α, IL‑4, IL‑5 and IL‑13) in bronchoalveolar lavage fluid. Further study found that SIRT3 overexpression significantly decreased apoptosis‑related protein expression (Bax/Bcl2 ratio and caspase 3 activity) and oxidative injury. In vitro, SIRT3 regulated oxidative stress‑induced bronchial epithelial cell (16HBE) apoptosis and cytokine expression. In conclusion, SIRT3 expression decreased in bronchial tissues of asthmatic mice and the upregulation of SIRT3 expression could reduce the apoptosis of bronchial epithelium and airway inflammation. It was concluded that SIRT3 might be a potential target in asthma treatment.

Introduction

Bronchial asthma (asthma) is the most common chronic respiratory disease in children. There are ~300 million individuals with asthma worldwide and the number of patients is increasing year by year (1). In China ~30 million individuals suffer from asthma and the prevalence rate is 1–4% (2). Asthma is a chronic inflammatory disease characterized by airway hyperresponsiveness, inflammatory cell infiltration, excessive mucus production and airway remodeling and it can be induced by respiratory tract infection, allergen exposure, strenuous exercise, climate change and other factors (3,4). Although the specific pathogenesis of asthma remains to be elucidated, airway inflammation is recognized as the most fundamental pathological change in its pathogenesis. Therefore, the inhibition of inflammation is an important strategy for asthma therapy. Glucocorticoids, among the most effective drugs for controlling airway inflammation, are widely used to treat asthma (3,4). However, long-term use of glucocorticoid treatment can cause a number of side effects, such as increased blood pressure, hyperglycemia, edema, osteoporosis, ulcer formation and weight gain (5). Therefore, it is essential to study the molecular mechanism of airway inflammation in asthma.

Airway epithelial cells are mainly composed of secretory and ciliated cells and they serve a critical role in the body's resistance to pathogenic microorganisms and the removal of foreign substances, such as dust particles (6,7). In asthmatic patients, the presence of airway inflammation leads to airway epithelial damage, including airway epithelial apoptosis, shedding and airway structure remodeling and these changes can weaken or cause the loss of the original function of the airway epithelium (8,9). Airway epithelium damage leads to the weakening of its physical barrier function, thereby increasing the risk of infection, forming a vicious circle that feeds on itself (8,9). Studying the apoptosis and inflammatory mechanisms of airway epithelial cells is therefore crucial for the treatment of asthma.

Sirtuins are a protein family that regulate cell health and serve a key role in regulating cell homeostasis (10). They are considered a new target for the treatment of bronchial inflammation (11). Sirtuin (SIRT)3 is a member of the sirtuin family that promotes longevity in a number of organisms, regulates inflammation through the modulation of mitochondrial function (12) and improves the antioxidant defense mechanisms (13,14). Although no studies of SIRT3 in asthma are reported, the sirtuin protein family serves an essential role in chronic obstructive pulmonary disease (15); SIRT3 can not only resist lung injury by resisting inflammation (16) and oxidation (17) but also protects bronchial epithelial cells by inhibiting oxidative stress (18) and autophagy (19). In addition, SIRT1, an upstream molecule that regulates the expression of SIRT3, is related to the severity of asthma (20,21). A study by Colley et al (22) also found that defective SIRT1 increases IL-4 expression through acetylation of GATA-3 in patients with severe asthma. Combined with the evidence by Liu et al (12) that SIRT1 suppresses inflammation by promoting SIRT3 expression, it was hypothesized that SIRT3 also served an essential role in asthma.

SIRT3 is also an apoptosis-related protein (23) and aberrant apoptosis of airway epithelial cells is also a contributing factor in development of asthma (24,25). Previous studies have found that downregulation of SIRT3 promotes the apoptosis of alveolar epithelial cells (26) and EC9706 cells (27). Studies have also shown that SIRT3 protects tumor cells against apoptosis under unfavorable environments, such as hypoxia (28), oxidized low-density lipoprotein (29) and high glucose levels (30). However, the expression of SIRT3 and its effect on apoptosis and bronchial inflammation in asthma remains to be elucidated. The present study attempted to investigate the role of SIRT3 in epithelial cells apoptosis induced by oxidative stress (H2O2) and the effect of SIRT3 overexpression on inflammation and airway epithelial structural integrity in asthmatic mice.

Materials and methods

Animals and experimental asthma model

The animal experiment was performed according to the protocols approved by the Beijing Luhe Hospital, Capital Medical University (approval no. 2021-LHKY-055-02). A total of 28 female C57BL/6 mice (age, 5–6 weeks; weight, 18–22 g) were used in the present study. Mice were purchased from Shanghai Lingchang Biological Technology Co., Ltd. and were kept at ~20-26°C, 40% relative humidity with 12 h light/dark cycles. After being housed under conventional conditions for one week prior to any experiments, 21 mice were used in the establishment of the asthma model, as illustrated in Fig. 1A (31). In brief, 200 µl of sensitizing fluid [containing 100 µg of ovalbumin (OVA) and 20 mg of Alum] was administered by intraperitoneal (i.p.) injection on day 0 and 14. From day 21, mice were given 40 ml of 2% OVA atomized solution through spray inhalation for 30 min/day for seven days. Control mice were given the same amount of saline solution at the same time. All mice were randomly distributed into 4 groups (7 mice/group): i) Control group; healthy mice; ii) Asthma group; experimental asthma mice model without any treatment; iii) Asthma + DEX group: experimental asthma mice model treated with 1 mg/Kg dexamethasone (DEX; cat. no. D4902; MilliporeSigma), i.p., for 7 days during the last week of OVA inhalation (32) and iv) Asthma + Ad-m-SIRT3 group; experimental asthma mice model infected with mice SIRT3 adenovirus (cat. no. AAV-272009; Vector BioLabs).

Lung resistance (LR) assessment and BALF collection

After 48 h from the last atomization with OVA, an AniRes2005 pulmonary function meter (Beijing Beilanbo Technology Co., Ltd.) was used to measure the lung resistance of mice to evaluate the experimental asthma model: Briefly, following intratracheal intubation, normal saline nebulization was used to determine basic lung resistance. After 2 min of rest, acetylcholine solutions of 50, 20, 10 and 5 mg/ml were nebulized for 2 min and lung resistance was subsequently determined. After assessing the lung function, the mice were euthanized by exposure to CO2 (20–30% of chamber volume per min) after being anesthetized with sodium pentobarbital (50 mg/kg i.p.) and the trachea was exposed. A 5 ml sterile syringe was then used to inject 2 ml of sterile PBS into the trachea and then the PBS solution was recovered. This process was repeated twice. The resultant collected cells in the PBS were bronchoalveolar lavage fluid (BALF). For cell count, cells were centrifuged at 500 × g for 5 min at room temperature for cell collection, resuspended in PBS and the cell suspension was smeared on a glass slide. Wright's Giemsa staining solution (cat. no. G3990; Beijing Solarbio Science & Technology Co., Ltd.) was used for cell staining for 1 min at room temperature and the cells were counted under an optical light microscope.

Infection of adenovirus in mice and 16HBE cells

After being anesthetized with sodium pentobarbital (50 mg/kg i.p.), 4×105 PFU/40 µl mice SIRT3 adenovirus (cat. no. AAV-272009, Vector Biolabs) was administered by nasal spray inhalation and the mice in the control group were infected with equivalent empty adenovirus, as previously described (33,34). After being infected with the adenovirus for 48 h, the mice were sacrificed to collect the bronchial tissues.

For adenovirus infection of 16HBE cells to knockdown or overexpress SIRT3, 1.0×106 16HBE cells were first seeded in a 6-well cell culture plate and cultured for 12 h at 37°C. To infect 16HBE cells 1×1010 PFU/100 µl SIRT3-AAV (cat. no. AAV-223017; Vector Biolabs) or SIRT3-short hairpin (sh)RNA adenovirus (cat. no. shAAV-272009; Vector Biolabs) were used to infect 16HBE cells and 16HBE cells were also infected with equivalent empty adenovirus [NC-shRNA (non-targeting sequence) (cat. no. shAAV-272009; Vector Biolabs) or NC-AAV (cat. no. shAAV-223017; Vector Biolabs)] as a negative control. After being infected with the adenovirus for 48 h at 37°C, the cells were collected for analysis.

Reverse transcription-quantitative (RT-q) PCR

An RNA extraction kit (cat. no. RC101-01, Vazyme Biotech Co., Ltd.) was used to extract the total RNA from cells (2×106) and tissues according to the manufacturer's protocol. Total RNA was reverse transcribed into cDNA using the PrimeScript RT reagent (cat. no. RR047A; Takara Bio, Inc.) according to the manufacturer's protocol. Next, 20 µl of RT-qPCR system was prepared, as described in the qPCR master mix kit instructions (cat. no. A6001; Promega Corporation) and the following thermocycling conditions were used: 95°C for 2 min; and 40 cycles at 95°C for 5 sec and 65°C for 15 sec. The relative expression of the genes was calculated using the 2−DDCq method (35). β-actin was used as a loading control. The primers used for qPCR analysis were: SIRT3-F: 5′-GGCTCTATACACAGAACATCGAC-3′, SIRT3-R: 5′-TAGCTGTTACAAAGGTCCCGT-3′; TNF-α-F: 5′-GACGTGGAACTGGCAGAAGAG-3′, TNF-α-R: 5′-TTGGTGGTTTGTGAGTGTGAG-3′; IL-4-F: 5′-ATCATCGGCATTTTGAACGAGG-3′, IL-4-R: 5′-TGCAGCTCCATGAGAACACTA-3′; IL-5-F: 5′-TCAGGGGCTAGACATACTGAAG-3′, IL-5-R: 5′-CCAAGGAACTCTTGCAGGTAAT-3′; IL-13-F: 5′-CAGCCTCCCCGATACCAAAAT-3′, IL-13-R: 5′-GCGAAACAGTTGCTTTGTGTAG-3′; β-actin-F: 5′-AGGGAAATCGTGCGTGACAT-3′, β-actin-F: 5′-GCCTCAGGAGTTTTGTCACCT-3′. These experiments were repeated three times.

Western blot analysis

Total protein was extracted from tissues using a Tissue Protein Extraction Reagent (cat. no. 78510; Thermo Fisher Scientific, Inc.) and a BCA kit (cat. no. 23227; Thermo Fisher Scientific, Inc.) was used to determine the protein concentration. Then, 50 µg total protein was analyzed using a 10% SDS-PAGE. After transferring to PVDF membranes (cat. no. LC2002; Thermo Fisher Scientific, Inc.) blocking was performed using 5% skimmed milk powder for 1 h at room temperature. The membranes were subsequently probed with primary antibodies against SIRT3 (1:1,000; cat. no. 2627; Cell Signaling Technology, Inc.), Bax (1:2,000; cat. no. ab32503; Abcam), Bcl2 (1:2,000; cat. no. ab196495; Abcam) and β-actin (1:4,000; cat. no. AF5001; Beyotime Institute of Biotechnology) at 4°C overnight. Subsequently, PVDF membranes were probed with goat anti-rabbit IgG heavy chain & light chain (H&L) HRP (1:2,000; cat. no. ab6721; Abcam) or goat anti-mouse IgG H&L HRP (1:2,000; cat. no. ab6789; Abcam) secondary antibodies for 1 h at room temperature. The proteins were visualized with an ECL solution (cat. no. WBKLS0100; Beijing Xinjingke Biotechnologies Co., Ltd.), followed by densitometry analysis using ImageJ v3.0 (National Institutes of Health). β-actin was used as control.

Immunohistochemistry staining of SIRT3

The expression of SIRT3 was detected in bronchial tissues using immunohistochemistry staining. Briefly, the bronchial tissues were harvested and fixed using 4% paraformaldehyde (cat. no. P1110; Beijing Solarbio Science & Technology Co., Ltd.) at 4°C overnight. Then histological paraffin sections (5 µm) were made after being embedded in paraffin and procedurally dehydrated (tissues were placed in 30, 50, 70, 80, 95 and 100% ethanol for 1 h at room temperature). There were seven mice in each group and ≥10 histological sections were prepared for each animal. The bronchial sections were incubated overnight at 4°C with the anti-SIRT3 antibody (1:500; cat. no. 2627; Cell Signaling Technology, Inc.). After a mild wash with PBS buffer, the bronchial sections were incubated with HRP-conjugated goat anti-rabbit antibody (1:2,000; cat. no. ab6721; Abcam) for 1 h at room temperature. The peroxidase activity was assessed via a 2 min reaction at room temperature with diaminobenzidine (DAB). The bronchial tissue sections were further counterstained with hematoxylin for 2 min at room temperature and images captured under a bright-field light microscope (Olympus BX51; Olympus Corporation).

TUNEL staining

To determine the apoptosis in the bronchial tissues, TUNEL staining was used as described previously (36). In brief, the mice bronchial tissue sections were fixed with 4% paraformaldehyde for 24 h at 4°C and were embedded in paraffin. Subsequently the samples were procedurally dehydrated (tissues were placed in 30, 50, 70, 80, 95 and 100% ethanol for 1 h at room temperature) and sectioned at 5 µm. After dewaxing and hydrating, tissue sections were incubated with proteinase K to permeate the cells for 30 min at 37°C. Next, the sections were incubated with the TUNEL reaction solution for 1 h at 37°C and then color development was achieved with DAB solution for 30 min at room temperature. The TUNEL Cell Apoptosis Detection kit (cat. no. TA201-02; TransGen Biotech Co., Ltd.) was used.

Caspase-3 activity measurement

Apoptosis was quantified by the caspase-3 activity measurement using the Ac-DEVD-7-Amino-4-trifluoromethylcoumarin (AFC) caspase-3 Fluorogenic Substrate (cat. no. 556574; BD Pharmingen; BD Biosciences) according to the manufacturer's instructions. Briefly, bronchial tissues from the control or asthma mice were lysed on ice. After centrifugation (12,000 × g for 10 min at 4°C), the supernatants were collected and the protein concentration was quantified using the BCA method. A total of 50 µg of proteins was pipetted and mixed with the assay buffer supplemented with 10 mM dithiothreitol (DTT). The fluorescence emission of the AFC (400 nm) was measured via the Spectra Max-Plus Microplate Spectrophotometer (Molecular Devices, LLC). The caspase-3 activity was expressed as nmol AFC/h/mg protein.

Detection of cytokines and serum immunoglobulin (Ig)E

The concentration of TNF-α, IL-4, IL-5 and IL-13 in bronchoalveolar lavage fluid (BALF) and serum IgE from mice were evaluated by ELISA kits as described in the manufacturer's instructions. The ELISA kits were: TNFα (cat. no. ab236712; Abcam), IL-4 (cat. no. ab100710; Abcam), IL-5 (cat. no. ab204523; Abcam), IL-13 (cat. no. ab100700; Abcam) and mice IgE (cat. no. ab157718; Abcam).

Detection of reactive oxygen species (ROS), glutathione (GSH), superoxide dismutase (SOD), glutathione peroxidase (Gpx) and malondialdehyde (MDA)

Homogenates were prepared with 50 mg of fresh bronchial tissue and the supernatant collected following bronchial tissue homogenate centrifugation (12,000 × g, 4°C, 10 min). Then, a ROS Detection kit (cat. no. S0058; Beyotime Institute of Biotechnology) was used to quantify the ROS levels. A GSH detection kit (cat. no. S0058; Beyotime Institute of Biotechnology) was used to detect the GSH levels. In addition, a SOD assay kit (cat. no. S0101S; Beyotime Institute of Biotechnology) was used to detect the SOD levels and an MDA assay kit (cat. no. S0101S; Beyotime Institute of Biotechnology) was used to detect the MDA levels. Finally, a Gpx assay kit (cat. no. S0058; Beyotime Institute of Biotechnology) was used to detect the levels of Gpx. All these assays were performed as according to the manufacturer's protocols.

Cell treatment and apoptosis assay

16HBE cells (1.5×106) were seeded in the 6-well cell culture plate and cultured for 12 h. After stimulating them with 0, 100, 200, or 400 µmol/l H2O2 for 12 h, the cells were collected and washed twice with cold-PBS buffer, then an Annexin V-PE/7-AAD Apoptosis Detection kit (cat. no. A213-01; Vazyme) was used to evaluate the apoptosis by flow cytometry (CytoFLEX S; Beckman Coulter, Inc.). In brief, cells were stained for 20 min at 4°C in the dark. Data were analyzed using FlowJo software (version 10.7.1; BD Biosciences) and the apoptotic rate was calculated as the percentage of early + late apoptotic cells.

Statistical analysis

Data were analyzed using SPSS v20.0 (IBM Corp.) and shown as mean ± SD. Student's t-test was used to compare the difference between two groups and one-way ANOVA followed by the Tukey's post hoc test was used to compare the difference between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

SIRT3 expression decreases in the bronchial tissues of the asthmatic mice

First, an asthma model was established through OVA sensitization induction, as represented in Fig. 1A. Symptoms such as sneezing, irritability, shortness of breath and scratching of the ears were observed in the asthma model mice after aerosolization. At the same time, DEX treatment could relieve the above symptoms, which indicated that the performance of the C57BL/6 as an asthma mice model after the challenge was consistent with the acute attack of asthma. Airway hyperresponsiveness (AHR) is a critical indicator of asthma and LR was used, which can reflect AHR to a certain extent, to represent AHR (37). As shown in Fig. 1B, acetylcholine increased mice LR in a dose-dependent manner and DEX reduced it. Similarly, the total content of IgE in the asthmatic mice serum was significantly higher when compared with the normal mice and DEX also significantly reduced it (Fig. 1C). Therefore, these results indicated that a mice asthma model had been successfully established.

Sirtuins, a group of class III deacetylases, have gained considerable attention for their positive effects on age-related diseases, such as cancer, cardiovascular disease, neurodegenerative diseases, osteoporosis and chronic obstructive pulmonary disease (15). SIRT3 serves an essential role in the sirtuin protein family and is also involved in mediating inflammation (14,38). As a disease with chronic airway inflammation as the primary clinical symptom, the present study found that the expression of SIRT3 mRNA was significantly lower in the bronchial tissues of asthmatic mice compared with the control (Fig. 2A). Similarly, the results of SIRT3 protein detection using western blotting (Fig. 2B and C) and immunochemistry (Fig. 2D) indicated that SIRT3 expression was significantly lower in bronchial tissues of asthmatic mice compared with the control. However, it was also found that DEX treatment could not change the decreased SIRT3 expression in the bronchial tissues of asthmatic mice (Fig. 2). Taken together, SIRT3 expression decreased in the bronchial tissues of asthmatic mice.

Upregulation of SIRT3 reduces the bronchial epithelial cell apoptosis in the asthmatic mice

As a mitochondrial protein, SIRT3-mediated mitophagy protects tumor cells against apoptosis under hypoxia (28) and the loss of SIRT3 promotes the apoptosis of EC9706 cells (27). This indicates that the loss of SIRT3 promotes apoptosis. It was hypothesized that the loss of SIRT3 was also involved in regulating the apoptosis of bronchial epithelial cells in asthmatic mice. To test this hypothesis, bronchial tissues were collected from the mice and apoptosis detected using TUNEL staining. It was found that the apoptotic (TUNEL positive) cells in bronchial tissue of asthmatic mice were significantly higher compared with the control mice (Fig. 3A and B). The results of the western blotting (Fig. 3C) demonstrated that Bax (a pro-apoptotic protein) protein expression levels in the asthma group were markedly higher compared with the control group, whereas Bcl2 (an anti-apoptotic protein) protein expression levels in the asthma group were markedly lower compared with the control group. SIRT3 protein expression levels were also markedly increased in the asthma group compared with the control group (Fig. 3D). The ratio of Bax/Bcl2 was significantly increased (Fig. 3E) in the bronchial tissues of asthmatic mice. Simultaneously, the caspase 3 activity in the bronchial tissues of asthmatic mice was significantly higher than in normal mice (Fig. 3F). To study the effect of SIRT3 expression on the apoptosis of bronchial epithelial cells in asthmatic mice, the expression of SIRT3 was upregulated using adenovirus overexpressing SIRT3 (Ad-m-SIRT3). It was found that Ad-m-SIRT3 successfully increased the expression of SIRT3 protein in bronchial tissues of asthmatic mice. Notably, overexpression of SIRT3 significantly reduced the increased TUNEL positive cells in bronchia from asthmatic mice and significantly reduced the increased Bax/Bcl2 ratio and caspase 3 activity in the bronchia from asthmatic mice (Fig. 3). In conclusion, upregulation of SIRT3 reduced bronchial epithelial cell apoptosis in asthmatic mice.

Inflammation in BALF of the asthmatic mice is suppressed by SIRT3 overexpression

SIRT3 is an essential molecule in inflammation regulation and was previously found to attenuate palmitate-induced ROS production and inflammation in proximal tubular cells (14). In the present study, the number of immune cells (macrophages, eosinophils, lymphocytes and neutrophils) in BALF from asthmatic mice increased significantly (Fig. 4A). The cytokines expression in BALF, such as TNF-α, IL-4, IL-5 and IL-13, was analyzed and it was found that the mRNAs of all these cytokines were also increased in BALF from asthmatic mice (Fig. 4B). Similarly, the ELISA results revealed that the content of TNF-α, IL-4, IL-5 and IL-13 in BALF from asthmatic mice were significantly higher compared with the BALF from normal mice (Fig. 4C). To study the effect of SIRT3 expression on BALA inflammation in asthmatic mice, the expression of SIRT3 was upregulated using adenovirus overexpressing SIRT3 (Ad-m-SIRT3). The results showed that overexpression of SIRT3 not only significantly reduced the increased number of immune cells (macrophages, eosinophils, lymphocytes and neutrophils) in BALF from asthmatic mice but also reduced the increased expression and content of cytokines from cells in BALF of asthmatic mice (Fig. 4). Together, these results suggested that the overexpression of SIRT3 reduced airway inflammation in asthmatic mice, which has the same effect as the specific drug DEX, which was used as a positive control, for the treatment of allergic rhinitis.

Upregulation of SIRT3 reduces the bronchial oxidative stress in the asthmatic mice

SIRT3 protects cells and inhibits inflammation by preventing oxidative stress in a number of diseases, such as diabetes (39), oral cancer (40) and myocardial ischemia-reperfusion (41). Oxidative stress serves an important role in the establishment of the asthma model by OVA sensitization induction (42) and oxidative stress can cause inflammation and apoptosis (43,44). Therefore, it was hypothesized that SIRT3 was involved in regulating airway inflammation and bronchial epithelial cell apoptosis by the modulation of bronchial oxidative stress in asthmatic mice. To test this hypothesis, the level of ROS was detected in the bronchial tissue by a ROS probe and it was found that the increased levels of ROS in the bronchial tissues of asthmatic mice were reduced by overexpression of SIRT3 (Fig. 5A). ELISA was used to observe the change in the endogenous antioxidants, GSH, SOD and Gpx, which are the antioxidants that help to reduce the content of ROS (45). The results of ELISA showed that the levels of GSH, SOD and Gpx in the bronchial tissues of asthmatic mice were all significantly lower compared with the control mice, while the decreased levels of GSH (Fig. 5B), SOD (Fig. 5C) and Gpx (Fig. 5D) in the bronchial tissues of asthmatic mice were reduced by overexpression of SIRT3. The levels of MDA, an indicator of the end products of the cellular membrane under oxidative injury, were detected and it was found that MDA levels increased in the bronchial tissues of asthmatic mice and overexpression of SIRT3 was able to reverse this (Fig. 5E). Therefore, these data suggested that the upregulation of SIRT3 reduced bronchial oxidative stress in the asthmatic mice.

SIRT3 regulated H2O2-induced apoptosis and inflammation in 16HBE cells

To study the relationship between SIRT3-regulated bronchial epithelial inflammation/apoptosis and oxidative stress, an oxidative stress cell model of bronchial epithelial cells (16HBE) was established, induced by H2O2 in vitro. Simultaneously, SIRT3 was knocked down through SIRT3-shRNA adenovirus infection and NC-shRNA adenovirus used as a negative control; SIRT3 was overexpressed through SIRT3-AAV adenovirus infection and the NC-AAV adenovirus used as a negative control. SIRT3-shRNA successfully knocked down SIRT3 expression and SIRT3-AAV successfully overexpressed SIRT3 in 16HBE cells (Fig. 6A). Subsequently, the induction concentration of H2O2 was analyzed and it was found that it induced the 16HBE cell apoptosis in a dose-dependent manner (Fig. 6B). The 100 µmol/l H2O2 dosage was chosen as the induction concentration since this concentration could induce a cell apoptosis rate of ~30%. After stimulating with 100 µmol/l H2O2 for 12 h, 16HBE cells were collected to analyze the apoptosis using flow cytometry. The results revealed that the knockdown of SIRT3 significantly increased H2O2-induced apoptosis and overexpression of SIRT3 significantly decreased H2O2-induced apoptosis in the 16HBE cells (Fig. 6C and D). Knockdown of SIRT3 significantly increased the H2O2-induced expression of cytokines (TNF-α, IL-4, IL-5 and IL-13) and the overexpression of SIRT3 significantly decreased the expression of these cytokines in 16HBE cells (Fig. 6E).

Discussion

The characteristics of an ideal asthma animal model include sudden onset of spasm, the occurrence of immediate and delayed allergies, airway hyperresponsiveness, chronic airway remodeling and lung function degradation (46,47). Previous studies have shown that OVA sensitization induction can establish this ideal asthma animal model (46,47). The present study established an asthma model through OVA sensitization and determined the expression of the SIRT3 gene by RT-qPCR, western blotting and immunochemistry. All the results suggested that SIRT3 expression was significantly decreased in the bronchia of asthma mice and was significantly associated with the pathologic features of asthma. Notably, upregulation of SIRT3 in the bronchial tissues could reduce the increased apoptosis of bronchial epithelial cells and significantly decrease the elevated airway inflammation in asthmatic mice. In addition, upregulation of SIRT3 reduced bronchial oxidative stress in asthmatic mice. In vitro, SIRT3 was found to regulate H2O2-induced apoptosis and inflammation in 16HBE cells. Asthma is the result of the interaction between genetic and environmental factors and it is a complex pathological process involving multiple gene regulations (9). However, chronic airway inflammation, abnormal apoptosis of airway epithelial cells, airway epithelial damage and repair disorders are the essential pathological basis of asthma (48,49). Therefore, studies on the pathogenesis of asthma, such as chronic airway inflammation and airway epithelial cell apoptosis, might find new targets for asthma treatment.

The SIRT3 gene is located on human chromosome 11 (11p15.5) and consists of 21,902 bases. SIRT3 protein is an evolutionarily highly conserved deacetylase whose activity depends on nicotinamide adenine dinucleotide and belongs to one of the proteins in the sirtuin family. In addition to regulating the body's energy metabolism, cell aging and tumor formation, SIRT3 also serves a pivotal role in regulating cell apoptosis through its enzymatic catalytic activity (23). However, to the best of the authors' knowledge, no data exist concerning the influence of SIRT3 on airway epithelial cell apoptosis. The present study found that the upregulation of SIRT3 in bronchial tissues could reduce the increased apoptosis of bronchial epithelial cells; however, the corresponding mechanism remains to be elucidated. Based on the antioxidant function of SIRT3 (50), it was hypothesized that SIRT3 might resist apoptosis by inhibiting oxidation. It was later found that the upregulation of SIRT3 reduced bronchial oxidative stress in asthmatic mice and reduced the H2O2-induced apoptosis in the 16HBE cells in vitro, while the loss of SIRT3 promoted H2O2-induced apoptosis in the 16HBE cells.

The body produces a large number of free radicals in metabolism and ROS are the main components. Under normal conditions, the level of ROS in the body maintains a dynamic balance, but this balance is broken by endogenous or exogenous harmful stimuli, resulting in a large amount of ROS accumulating in the body, destroying the balance between the oxidative system and the antioxidant system, developing oxidative stress and causing cell oxidative damage or apoptosis (51,52). It has been confirmed that oxidative stress is closely related to cell apoptosis and ROS serves a vital role in this process (51,52). A previous study has shown that ROS can activate nuclear transcription factor NF-κB leading to cell apoptosis by activating proteinase C (53). ROS can also activate the P53-induced apoptosis pathway by damaging DNA (54) and can also lead to cell apoptosis by damaging mitochondria and initiating the mitochondrial-mediated apoptosis pathways (39). In brief, inhibiting the generation of ROS and/or increasing the body's ability to eliminate it will inhibit cell apoptosis to some extent. Notably, SIRT3 is closely related to oxidative stress: Fu et al (50) and Zhang et al (55) confirmed that the activity of Mn-SOD in the liver of SIRT3 knockout mice is reduced and SIRT3 can significantly enhance the activity of Mn-SOD by reducing the acetylation levels of K53 and K89 or by deacetylating the 122 lysine group of Mn-SOD, thereby reducing the ROS levels in the cells.

It is hypothesized that in the pathogenesis of asthma, the ratio or functional imbalance of T helper (Th)1 and Th2 cells, especially the increase of Th2 cells hyperfunction, are important immunological abnormal factors (56). Th2 cells mainly secrete IL-4, IL-5, IL-13 and other cytokines and mediate the occurrence of asthma through multiple pathways, such as efficient recruitment and activation of eosinophils, stimulation of Ig conversion to produce IgE and others (57). TNF-α is a pro-inflammatory cytokine that participates in the immune and inflammatory responses and can mediate the elevation of several inflammatory cell numbers such as eosinophils and neutrophils, generate a variety of inflammatory mediators and aggravate the inflammatory response (58,59). TNF-α can also stimulate endothelial cells and macrophages to release IL-6 and other inflammatory mediators, causing airway inflammation, destroying collagen networks and participating in airway remodeling (60,61). In asthma, TNF-α expression in the airways and lungs is significantly increased, promoting the occurrence and development of inflammatory reactions (60,61). Inflammation and oxidative stress are not independent of each other (62,63): Inflammation can cause oxidative stress and oxidative stress can also cause inflammation, including oxidative stress-induced airway inflammation (64). In a number of previous studies, a cell oxidative stress induced by H2O2 model was used to study inflammation, such as Cao et al (65) and de Oliveira-Marques et al (66), including H2O2-induce 16HBE cells (67). The results of the present study suggested that the upregulation of SIRT3 could decrease the elevated airway inflammation in asthmatic mice and reduce the H2O2-induced inflammation in the 16HBE cells in vitro, while the loss of SIRT3 improved the H2O2-induced inflammation in the 16HBE cells.

To the best of the authors' knowledge, no relation has been established between SIRT3 and inflammation in the bronchial tissue in asthma. However, the study of SIRT3 inhibiting inflammation has been widely reported: Koyama et al (14) found that SIRT3 attenuates palmitate-induced ROS production and inflammation in proximal tubular cells and Boniakowski et al (38) describes that SIRT3 suppresses macrophage-mediated inflammation in diabetic wound repair. The findings of the present study shared broad similarities with an earlier study by Kim et al (68) who report that KIF3A deficiency in mice results in increased inflammation. Thus, it was hypothesized that decreased expression of SIRT3 in the bronchial epithelium of asthmatic animals may modulate the airway inflammation, which needs further validation and support. In conclusion, the results of the present study indicated that the SIRT3 gene may be a key molecule for targeted therapeutics for asthma. However, the lack of data on SIRT3 expression in clinical studies is limited and therefore needs to be explored in future work.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JS conceived the study, wrote the manuscript, performed the data analysis, participated in the experiments and data collection. JW designed and supervised the study and edited the manuscript. JS and JW confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Beijing Luhe Hospital, Capital Medical University (Tongzhou, China; approval no. 2021-LHKY-055-02).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Boulet LP, Reddel HK, Bateman E, Pedersen S, FitzGerald JM and O'Byrne PM: The global initiative for asthma (GINA): 25 years later. Eur Respir J. 54:19005982019. View Article : Google Scholar : PubMed/NCBI

2 

Lin J, Wang W, Chen P, Zhou X, Wan H, Yin K, Ma L, Wu C, Li J, Liu C, et al: Prevalence and risk factors of asthma in mainland China: The CARE study. Respir Med. 137:48–54. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Lambrecht BN, Hammad H and Fahy JV: The cytokines of asthma. Immunity. 50:975–991. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Pavord ID, Beasley R, Agusti A, Anderson GP, Bel E, Brusselle G, Cullinan P, Custovic A, Ducharme FM, Fahy JV, et al: After asthma: Redefining airways diseases. Lancet. 391:350–400. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Heffler E, Madeira LNG, Ferrando M, Puggioni F, Racca F, Malvezzi L, Passalacqua G and Canonica GW: Inhaled corticosteroids safety and adverse effects in patients with asthma. J Allergy Clin Immunol Pract. 6:776–781. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Kato A and Schleimer RP: Beyond inflammation: Airway epithelial cells are at the interface of innate and adaptive immunity. Curr Opin Immunol. 19:711–720. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Hiemstra PS, McCray PB Jr and Bals R: The innate immune function of airway epithelial cells in inflammatory lung disease. Eur Respir J. 45:1150–1162. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Lopez-Guisa JM, Powers C, File D, Cochrane E, Jimenez N and Debley JS: Airway epithelial cells from asthmatic children differentially express proremodeling factors. J Allergy Clin Immunol. 129:990–997.e6. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Haddad A, Gaudet M, Plesa M, Allakhverdi Z, Mogas AK, Audusseau S, Baglole CJ, Eidelman DH, Olivenstein R, Ludwig MS and Hamid Q: Neutrophils from severe asthmatic patients induce epithelial to mesenchymal transition in healthy bronchial epithelial cells. Respir Res. 20:2342019. View Article : Google Scholar : PubMed/NCBI

10 

Wątroba M, Dudek I, Skoda M, Stangret A, Rzodkiewicz P and Szukiewicz D: Sirtuins, epigenetics and longevity. Ageing Res Rev. 40:11–19. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Ma K, Lu N, Zou F and Meng FZ: Sirtuins as novel targets in the pathogenesis of airway inflammation in bronchial asthma. Eur J Pharmacol. 865:1726702019. View Article : Google Scholar : PubMed/NCBI

12 

Liu TF, Vachharajani V, Millet P, Bharadwaj MS, Molina AJ and Mccall CE: Sequential actions of SIRT1-RELB-SIRT3 coordinate nuclear-mitochondrial communication during immunometabolic adaptation to acute inflammation and sepsis. J Biol Chem. 290:396–408. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Ren JH, Xiang C, Li Z, Tao NN, Zhou HZ, Liu B, Li WY, Huang AL and Chen J: Protective role of sirtuin3 (SIRT3) in Oxidative stress mediated by hepatitis B virus X protein expression. PLoS One. 11:e01509612016. View Article : Google Scholar : PubMed/NCBI

14 

Koyama T, Kume S, Koya D, Araki S, Isshiki K, Chin-Kanasaki M, Sugimoto T, Haneda M, Sugaya T, Kashiwagi A, et al: SIRT3 attenuates palmitate-induced ROS production and inflammation in proximal tubular cells. Free Radic Biol Med. 51:1258–1267. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Chun P: Role of sirtuins in chronic obstructive pulmonary disease. Arch Pharm Res. 38:1–10. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Kurundkar D, Kurundkar AR, Bone NB, Becker EJ Jr, Liu W, Chacko B, Darley-Usmar V, Zmijewski JW and Thannickal VJ: SIRT3 diminishes inflammation and mitigates endotoxin-induced acute lung injury. JCI Insight. 4:e1207222019. View Article : Google Scholar : PubMed/NCBI

17 

Tian YG and Zhang J: Protective effect of SIRT3 on acute lung injury by increasing manganese superoxide dismutase-mediated antioxidation. Mol Med Rep. 17:5557–5565. 2018.PubMed/NCBI

18 

Zhang M, Zhang Y, Roth M, Zhang L, Shi R, Yang X, Li Y and Zhang J: Sirtuin 3 inhibits airway epithelial mitochondrial oxidative stress in cigarette smoke-induced COPD. Oxid Med Cell Longev. 2020:75829802020. View Article : Google Scholar : PubMed/NCBI

19 

Chen IC, Huang HH, Chen PF and Chiang HC: Sirtuin 3 protects against urban particulate matter-induced autophagy in human bronchial epithelial cells. Toxicol Sci. 152:113–127. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Wang Y, Li D, Ma G, Li W, Wu J, Lai T, Huang D, Zhao X, Lv Q, Chen M and Wu B: Increases in peripheral SIRT 1: A new biological characteristic of asthma. Respirology. 20:1066–1072. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Tsilogianni Z, Baker JR, Papaporfyriou A, Papaioannou AI, Papathanasiou E, Koulouris NG, Daly L, Ito K, Hillas G, Papiris S, et al: Sirtuin 1: Endocan and sestrin 2 in different biological samples in patients with asthma. Does severity make the difference? J Clin Med. 9:4732020.PubMed/NCBI

22 

Colley T, Mercado N, Kunori Y, Brightling C, Bhavsar PK, Barnes PJ and Ito K: Defective sirtuin-1 increases IL-4 expression through acetylation of GATA-3 in patients with severe asthma. J Allergy Clin Immunol. 137:1595–1597. –e7. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Giralt A and Villarroya F: SIRT3, a pivotal actor in mitochondrial functions: Metabolism, cell death and aging. Biochem J. 444:1–10. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Roscioli E, Hamon R, Ruffin RE, Lester S and Zalewski P: Cellular inhibitor of apoptosis-2 is a critical regulator of apoptosis in airway epithelial cells treated with asthma-related inflammatory cytokines. Physiol Rep. 1:e001232013. View Article : Google Scholar : PubMed/NCBI

25 

Trautmann A, Schmid-Grendelmeier P, Krüger K, Crameri R, Akdis M, Akkaya A, Bröcker EB, Blaser K and Akdis CA: T cells and eosinophils cooperate in the induction of bronchial epithelial cell apoptosis in asthma. J Allergy Clin Immunol. 109:329–337. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Jablonski RP, Kim SJ, Cheresh P, Williams DB, Morales-Nebreda L, Cheng Y, Yeldandi A, Bhorade S, Pardo A, Selman M, et al: SIRT3 deficiency promotes lung fibrosis by augmenting alveolar epithelial cell mitochondrial DNA damage and apoptosis. FASEB J. 31:2520–2532. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Yang M, Yang C and Pei Y: Effects of downregulation of SIRT3 expression on proliferation and apoptosis in esophageal squamous cell carcinoma EC9706 cells and its molecular mechanisms. Biomed Mater Eng. 24:3883–3890. 2014.PubMed/NCBI

28 

Qiao A, Wang K, Yuan Y, Guan Y, Ren X, Li L, Chen X, Li F, Chen AF, Zhou J, et al: Sirt3-mediated mitophagy protects tumor cells against apoptosis under hypoxia. Oncotarget. 7:43390–43400. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Luo X, Yang Z, Zheng S, Cao Y and Wu Y: Retracted: Sirt3 activation attenuated oxidized low-density lipoprotein-induced human umbilical vein endothelial cells' apoptosis by sustaining autophagy. Cell Biol Int. 41:9322017. View Article : Google Scholar : PubMed/NCBI

30 

Jiao X, Li Y, Zhang T, Liu M and Chi Y: Role of Sirtuin3 in high glucose-induced apoptosis in renal tubular epithelial cells. Biochem Biophys Res Commun. 480:387–393. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Geng G, Du Y, Dai J, Tian D, Xia Y and Fu Z: KIF3A knockdown sensitizes bronchial epithelia to apoptosis and aggravates airway inflammation in asthma. Biomed Pharmacother. 97:1349–1355. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Yilmaz O, Karaman M, Bagriyanik HA, Firinci F, Kiray M, Turkeli A, Karaman O and Yuksel H: Comparison of TNF antagonism by etanercept and dexamethasone on airway epithelium and remodeling in an experimental model of asthma. Int Immunopharmacol. 17:768–773. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Yang C, Li J, Lin H, Zhao K and Zheng C: Nasal mucosa derived-mesenchymal stem cells from mice reduce inflammation via modulating immune responses. PLoS One. 10:e01188492015. View Article : Google Scholar : PubMed/NCBI

34 

Karra L, Haworth O, Priluck R, Levy BD and Levi-Schaffer F: Lipoxin B4 promotes the resolution of allergic inflammation in the upper and lower airways of mice. Mucosal Immunol. 8:852–862. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Hu J, Zhang G and Selzer ME: Activated caspase detection in living tissue combined with subsequent retrograde labeling, immunohistochemistry or in situ hybridization in whole-mounted lamprey brains. J Neurosci Methods. 220:92–98. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Wang H, Liu Y, Shi J and Cheng Z: ORMDL3 knockdown in the lungs alleviates airway inflammation and airway remodeling in asthmatic mice via JNK1/2-MMP-9 pathway. Biochem Biophys Res Commun. 516:739–746. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Boniakowski AE, denDekker AD, Davis FM, Joshi A, Kimball AS, Schaller M, Allen R, Bermick J, Nycz D, Skinner ME, et al: SIRT3 regulates macrophage-mediated inflammation in diabetic wound repair. J Invest Dermatol. 139:2528–2537.e2. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Zheng J, Shi L, Feng L, Xu W, Li T, Gao L, Sun Z, Yu J and Zhang J: Sirt3 ameliorates oxidative stress and mitochondrial dysfunction after intracerebral hemorrhage in diabetic rats. Front Neurosci. 12:4142018. View Article : Google Scholar : PubMed/NCBI

40 

Kao YY, Chou CH, Yeh LY, Chen YF, Chang KW, Liu CJ, Fan Chiang CY and Lin SC: MicroRNA miR-31 targets SIRT3 to disrupt mitochondrial activity and increase oxidative stress in oral carcinoma. Cancer Lett. 456:40–48. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Chang G, Chen Y, Zhang H and Zhou W: Trans sodium crocetinate alleviates ischemia/reperfusion-induced myocardial oxidative stress and apoptosis via the SIRT3/FOXO3a/SOD2 signaling pathway. Int Immunopharmacol. 71:361–371. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Sahiner UM, Birben E, Erzurum S, Sackesen C and Kalayci Ö: Oxidative stress in asthma: Part of the puzzle. Pediatr Allergy Immunol. 29:789–800. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Mishra V, Banga J and Silveyra P: Oxidative stress and cellular pathways of asthma and inflammation: Therapeutic strategies and pharmacological targets. Pharmacol Ther. 181:169–182. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Chandra J, Samali A and Orrenius S: Triggering and modulation of apoptosis by oxidative stress. Free Radic Biol Med. 29:323–333. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Pisoschi AM and Pop A: The role of antioxidants in the chemistry of oxidative stress: A review. Eur J Med Chem. 97:55–74. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Yuk JE, Lee MY, Kwon OK, Cai XF, Jang HY, Oh SR, Lee HK and Ahn KS: Effects of astilbic acid on airway hyperresponsiveness and inflammation in a mouse model of allergic asthma. Int Immunopharmacol. 11:266–273. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Ji W, Chen X, Zhengrong C, Yumin H, Huang L and Qiu Y: Therapeutic effects of anti-B7-1 antibody in an ovalbumin-induced mouse asthma model. Int Immunopharmacol. 8:1190–1195. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Xie CH, Cao YM, Huang Y, Shi QW, Guo JH, Fan ZW, Li JG, Chen BW and Wu BY: Long non-coding RNA TUG1 contributes to tumorigenesis of human osteosarcoma by sponging miR-9-5p and regulating POU2F1 expression. Tumour Biol. 37:15031–15041. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Marszalek JR, Liu X, Roberts EA, Chui D, Marth JD, Williams DS and Goldstein LS: Genetic evidence for selective transport of opsin and arrestin by kinesin-II in mammalian photoreceptors. Cell. 102:175–187. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Fu Y, Kinter M, Hudson J, Humphries KM, Lane RS, White JR, Hakim M, Pan Y, Verdin E and Griffin TM: Aging promotes sirtuin 3-dependent cartilage superoxide dismutase 2 acetylation and osteoarthritis. Arthritis Rheumatol. 68:1887–1898. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Loh KP, Huang SH, De Silva R, Tan BK and Zhu YZ: Oxidative stress: Apoptosis in neuronal injury. Curr Alzheimer Res. 3:327–337. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Lushchak VI: Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact. 224:164–175. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Yijing GF and Tangxue-Ming SY: Activation of Transcription Factor NF-kB and Accumulation of Reactive Oxygen Species Are Involved in Arsenic Trioxide-induced Apoptosis of Esophageal. Carcinoma Cells. 5:140. 2000.(In Chinese).

54 

Ye J, Wang S, Leonard SS, Sun Y, Butterworth L, Antonini J, Ding M, Rojanasakul Y, Vallyathan V, Castranova V and Shi X: Role of reactive oxygen species and p53 in chromium(VI)-induced apoptosis. J Biol Chem. 274:34974–34980. 1999. View Article : Google Scholar : PubMed/NCBI

55 

Zhang J, Song X, Cao W, Lu J, Wang X, Wang G, Wang Z and Chen X: Autophagy and mitochondrial dysfunction in adjuvant-arthritis rats treatment with resveratrol. Sci Rep. 6:329282016. View Article : Google Scholar : PubMed/NCBI

56 

Riiser A: The human microbiome, asthma and allergy. Allergy Asthma Clin Immunol. 11:352015. View Article : Google Scholar : PubMed/NCBI

57 

Fahy JV: Type 2 inflammation in asthma-present in most, absent in many. Nat Rev Immunol. 15:57–65. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Almishri W, Santodomingo-Garzon T, Le T, Stack D, Mody CH and Swain MG: TNFα augments cytokine-induced NK cell IFNγ production through TNFR2. J Innate Immun. 8:617–629. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Lampropoulou IT, Stangou Μ, Sarafidis P, Gouliovaki A, Giamalis P, Tsouchnikas I, Didangelos T and Papagianni Α: TNF-α pathway and T-cell immunity are activated early during the development of diabetic nephropathy in type II diabetes mellitus. Clin Immunol. 215:1084232020. View Article : Google Scholar : PubMed/NCBI

60 

Brightling C, Berry M and Amrani Y: Targeting TNF-alpha: A novel therapeutic approach for asthma. J Allergy Clin Immunol. 121:5–10; quiz 11–2. 2008. View Article : Google Scholar : PubMed/NCBI

61 

Berry M, Brightling C, Pavord I and Wardlaw A: TNF-alpha in asthma. Curr Opin Pharmacol. 7:279–282. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Tabet F and Rye KA: High-density lipoproteins, inflammation and oxidative stress. Clin Sci (Lond). 116:87–98. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Yoshikawa T: Inflammation and oxidative stress. Nihon Naika Gakkai Zasshi. 95:1870–1875. 2006.(In Japanese). View Article : Google Scholar : PubMed/NCBI

64 

Chen Q, Zhou Y, Zhou L, Fu Z, Yang C, Zhao L, Li S, Chen Y, Wu Y, Ling Z, et al: TRPC6-dependent Ca2+ signaling mediates airway inflammation in response to oxidative stress via ERK pathway. Cell Death Dis. 11:1702020. View Article : Google Scholar : PubMed/NCBI

65 

Cao YJ, Zhang YM, Qi JP, Liu R, Zhang H and He LC: Ferulic acid inhibits H2O2-induced oxidative stress and inflammation in rat vascular smooth muscle cells via inhibition of the NADPH oxidase and NF-κB pathway. Int Immunopharmacol. 28:1018–1025. 2015. View Article : Google Scholar : PubMed/NCBI

66 

de Oliveira-Marques V, Cyrne L, Marinho HS and Antunes F: A quantitative study of NF-kappaB activation by H2O2: Relevance in inflammation and synergy with TNF-alpha. J Immunol. 178:3893–3902. 2007. View Article : Google Scholar : PubMed/NCBI

67 

Suwara MI, Borthwick LA, Mann J, Fisher AJ and Mann DA: Inflammation: An important Regulator of the Fibrotic Response: S120 IL-1 is a Key Epithelial Alarmin Which Oromotes Fibroblast Activation. Newcastle University; Newcastle: 2010

68 

Kim D, Park W, Lee S, Kim W, Park SK and Kang KP: Absence of Sirt3 aggravates cisplatin nephrotoxicity via enhanced renal tubular apoptosis and inflammation. Mol Med Rep. 18:3665–3672. 2018.PubMed/NCBI

Related Articles

Journal Cover

April-2022
Volume 25 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song J and Song J: SIRT3 regulates bronchial epithelium apoptosis and aggravates airway inflammation in asthma. Mol Med Rep 25: 144, 2022
APA
Song, J., & Song, J. (2022). SIRT3 regulates bronchial epithelium apoptosis and aggravates airway inflammation in asthma. Molecular Medicine Reports, 25, 144. https://doi.org/10.3892/mmr.2022.12660
MLA
Song, J., Wang, J."SIRT3 regulates bronchial epithelium apoptosis and aggravates airway inflammation in asthma". Molecular Medicine Reports 25.4 (2022): 144.
Chicago
Song, J., Wang, J."SIRT3 regulates bronchial epithelium apoptosis and aggravates airway inflammation in asthma". Molecular Medicine Reports 25, no. 4 (2022): 144. https://doi.org/10.3892/mmr.2022.12660