Tremella polysaccharides inhibit cellular apoptosis and autophagy induced by Pseudomonas aeruginosa lipopolysaccharide in A549 cells through sirtuin 1 activation

Tremella

Abstract

In the present study, the role of Tremella polysaccharides in cellular apoptosis and autophagy induced by Pseudomonas aeruginosa lipopolysaccharide (LPS) in human epithelial A549 lung-cancer cells was investigated. Initially, it was demonstrated that LPS attenuated A549 cell viability in a time- and dose-dependent manner. Furthermore, LPS induced apoptotic cell death and autophagy in A549 cells and increased reactive oxygen species (ROS) production in a time-dependent manner. In addition, LPS treatment was demonstrated to markedly suppress sirtuin 1 (SIRT1) protein expression in A549 cells. Notably, it was demonstrated that Tremella polysaccharides activate SIRT1, leading to increased p62 expression, decreased p53 acetylation and B-cell lymphoma 2-associated X protein expression, and subsequently attenuate LPS-induced apoptotic cell death and autophagy. The results of the present study demonstrated that Tremella polysaccharides activate SIRT1 and inhibit LPS-induced ROS production, apoptosis and autophagy. This may have critical implications for the treatment of Pseudomonas aeruginosa infection.

Introduction

Pseudomonas aeruginosa is a prevalent opportunistic and virulent pathogen in pediatric patients (1,2). It is a common Gram-negative bacterium, which produces a variety of virulence factors, including exotoxins and enzymes. The major virulence factor of P. aeruginosa is lipopolysaccharide (LPS) (3,4). LPS is a major cause of respiratory tract disease in infants and young children worldwide. Therefore, it is crucial to develop effective preventative measures for LPS infection in pediatric patients. It has been demonstrated that LPS induces apoptotic cell death and intracellular reactive oxygen species (ROS) generation (5,6). ROS generation induces oxidative stress-mediated apoptotic cell death (7,8). Sirtuin 1 (SIRT1) is a nicotinamide-adenine dinucleotide-dependent class III protein deacetylase that belongs to the silent information regulator 2 gene family (7–10). SIRT1 has been associated with several physiological processes, including cellular apoptosis, autophagy, endocrine signaling, metabolism and chromatin remodeling (9–11). Tremella polysaccharides (TP) are isolated from the fruiting bodies and silver cell spore fermentation in Tremella fungi (12). Several biological actions have been attributed to TP, including cytokine-stimulation, anti-inflammatory and anti-diabetic activities (13,14). In a murine model, treatment with TP suppressed cancer cell DNA synthesis and growth (15). However, little is known about the anti-inflammatory role of TP in LPS infection in lung cancer.

In the present study, the role of TP in LPS-induced apoptosis and autophagy in A549 lung cancer cells was investigated. It was demonstrated that LPS suppresses SIRT1 protein expression, whereas treatment with TP increases SIRT1 expression and subsequently inhibits LPS-induced apoptosis and autophagy in A549 lung cancer cells.

Materials and methods

Cell culture and treatments 

A549 lung cancer cells (American Type Culture Collection, Manassas, VA, USA) were maintained in DMEM/F-12 (HyClone; GE Healthcare Life Sciences, Logan, UT, USA), supplemented with 10% heat-inactivated fetal bovine serum (HyClone; GE Healthcare Life Sciences) and 100 U/ml penicillin and streptomycin. Cells were grown in 25 cm2 culture flasks at 37°C in a humidified atmosphere containing 5% CO2. LPS (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was dissolved in PBS. Pretreatment with 10 µg/ml TP [dissolved in dimethyl sulfoxide (DMSO)] (Sigma-Aldrich; Merck KGaA) lasted for 48 h, and then there was an interval of 1 h prior to LPS treatment.

Cell viability assay 

Cell viability was determined using the colorimetric MTT assay (Sigma-Aldrich; Merck KGaA). Briefly, A549 cells were seeded in 96-well tissue culture plates at a density of 5×104 cells/well. After 24 h, cells were treated with 0.5, 1 or 10 µg/ml LPS for 48 h or 10 µg/ml LPS for 12, 24 or 48 h, and then cultured in fresh medium containing 0.5 mg/ml MTT for 4 h at 37°C. Subsequently, the formazan crystals that formed were dissolved in DMSO and the absorbance was determined at 550 nm.

Intracellular ROS quantification 

A549 cells were seeded in 6-well tissue culture plates at a density of 1×105 cells/well. When they had reached 70–80% confluence, cells were cultured for 16 h in serum-free DMEM/F-12. Following treatment with 10 µg/ml LPS for 48 h at 37°C, cells were incubated with 1 mM dichlorodihydrofluorescein diacetate for 40 min at 37°C in the dark. Cells were harvested and re-suspended in PBS. The relative fluorescence intensity was determined using a flow cytometer a BD FACSCalibur system (BD Biosciences, Franklin Lakes, NJ, USA) and data was analyzed using the ModFit software version 4.1 (Verity Software House, Inc., Topsham, ME, USA).

Flow cytometric analysis 

Cellular apoptosis was determined using the Annexin V-Fluorescein Isothiocyanate (FITC) Apoptosis Detection kit (BD Biosciences), according to the manufacturer’s protocol. Following treatment with 10 µg/ml LPS for 48 h at 37°C, cells were collected in a 5 ml culture tube and washed twice with ice-cold PBS. Cells were subsequently re-suspended in binding buffer and transferred to a new 5 ml culture tube. Annexin V-FITC (5 µl) and propidium iodide (5 µl) were added, and cells were incubated at room temperature for 15 min in the dark. Finally, 400 µl binding buffer was added and apoptotic cells were analyzed using a FC500 flow cytometry instrument equipped with CXP 2.0 software (Beckman Coulter, Bethesda, MA, USA) within 1 h.

Hoechst 33258 staining 

Following treatment with 10 µg/ml LPS for 48 h at 37°C, cells were washed with ice-cold PBS three times and then fixed with 4% formaldehyde in PBS for 15 min (37°C). Subsequently, cells were stained with Hoechst 33258 (10 µg/ml) at 37°C for 5 min and washed three times with PBS. Finally, cells were observed under a fluorescent microscope (Olympus Corporation, Tokyo, Japan; magnification, ×40).

Western blotting 

Total protein was extracted from cells using radioimmunoprecipitation assay buffer (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China), according to the manufacturer’s protocol. A bicinchoninic protein assay kit (Pierce; Thermo Fisher Scientific, Inc.) was used to determine the protein concentration. Immunoblotting was performed as previously described (16). Primary antibodies against SIRT1 (cat. no., 9475; dilution, 1:1,000), p62 (cat. no., 23214; dilution, 1:1,000), acetyl-p53 (cat. no., 2570; dilution, 1:1,000), p53 (cat. no., 2524; dilution, 1:1,000), B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax; cat. no., 5023; dilution, 1:1,000), Bcl-2 (cat. no., 2872; dilution, 1:1,000), and β-actin (cat. no., 4970; dilution, 1:4,000) were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA). These antibodies were incubated with the membranes at 4°C overnight. Following three washes (10 min/wash) with TBST, the membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG (1:5,000; cat. no., ZB-2306; Zhongshan Golden Bridge Biological Technology Co., Beijing, China) for 2 h at room temperature, and then washed three times (10 min/wash). The proteins were detected using enhanced chemiluminescence, according to the manufacturer’s protocol (Merck KGaA, Darmstadt, Germany). ImageJ 1.8.0 (National Institutes of Health, Bethesda, MD, USA) was applied to quantify the relative protein levels. β-actin was used as an internal control.

Transient transfection of green fluorescent protein-microtubule-associated protein 1A/1B-light chain 3 (GFP-LC3)-expressing plasmid 

A549 cells were seeded at a density of 5×105 cells/well in 6-well plates for 24 h. Transfection of a GFP-LC3-expressing plasmid (cat. no. 17-10230; Merck KGaA, Darmstadt, Germany) was performed using Lipofectamine 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA), according to the manufacturer’s protocol. After 24 h, cells were treated with 10 µg/ml LPS or 10 ng/ml rapamycin (Rap; R8718, Sigma-Aldrich; Merck KGaA) for 48 h at 37°C, and incubated for 16 h. GFP-LC3-positive cells were observed using a confocal laser microscope (LSM700; Carl Zeiss AG, Oberkochen, Germany).

Electron microscopy 

Samples were fixed with 2% glutaraldehyde-paraformaldehyde in 0.1 M phosphate buffer, pH 7.4, for 12 h at 4°C and washed three times for 30 min with 0.1 M phosphate buffer. Subsequently, samples were incubated with 1% OsO4 dissolved in 0.1 M phosphate buffer for 2 h, dehydrated in an ascending ethanol series (50–100%) and infiltrated with propylene oxide. The Poly/Bed® 812 kit (Polysciences, Inc., Warrington, PA, USA) was used for resin embedding and polymerization was performed at 60°C in an electron microscope oven (TD-700; DOSAKA, Kyoto, Japan) for 24 h according to the manufacturer’s protocol. Sections 350 nm in size were cut and stained using toluidine blue for 20 min at room temperature (for light microscopy) and 70 nm thin sections were double-stained using 7% uranyl acetate and lead citrate for 20 min at room temperature. Sections were cut using a Leica Ultracut UCT Ultra-microtome (Leica Microsystems GmbH, Wetzlar, Germany) and observed using a transmission electron microscope (JEM-1011; JEOL, Ltd., Tokyo, Japan) at an acceleration voltage of 80 kV.

Transient transfection 

Cells were transfected with small interfering RNA (siRNA) targeting si-SIRT1 (12241, Cell Signaling Technology, Inc., Danvers, MA, USA), or with negative control siRNA (NC; 5′-ACUAGUCGAUCUAUGUGUGAUATT-3′) (Shanghai GenePharma Co., Ltd., Shanghai, China) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at the final concentration of 20 nM, according to the manufacturer’s protocol. In brief, A549 cells (1×106 cells/well) were seeded in a 6-well plate with 2 ml RPMI-1640 medium (HyClone; GE Healthcare Life Sciences). At 60% confluence, si-SIRT1 or NC was mixed with Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature for 20 min. Then, the mixture was added into each well at a final concentration of 20 nM for 48 h. Then, the cells were collected for further analysis.

Statistical analysis 

Data are expressed as the mean ± standard error of the mean. Multiple comparisons were evaluated by analysis of variance followed by Tukey’s multiple-comparison test. P<0.05 was considered to indicate a statistically significant difference.

Results

LPS inhibits the viability of A549 cells in a time- and dose-dependent manner 

A549 cells were treated with 0.5, 1 or 10 µg/ml LPS for 48 h, and cell viability was determined using the MTT assay. As presented in Fig. 1A, treatment with LPS suppressed the viability of A549 cells in a dose-dependent manner. A549 cells were also treated with 10 µg/ml LPS for 12, 24, 48 h. Notably, treatment with LPS inhibited the viability of A529 cells in a dose-dependent manner (Fig. 1B).

 
An external file that holds a picture, illustration, etc.
Object name is ol-15-06-9609-g00.jpg

LPS inhibits the viability of A549 cells in a time- and dose-dependent manner. (A) MTT assay of A549 cells treated with 0.5, 1 or 10 µg/ml LPS for 48 h. (B) MTT assay of A549 cells treated with 10 µg/ml LPS for 12, 24 or 48 h. *P<0.05, **P<0.01 vs. the untreated control. LPS, Pseudomonas aeruginosa lipopolysaccharide; OD, optical density; DMSO, dimethyl sulfoxide.

LPS induces apoptosis in A549 cells 

The effect of LPS treatment on the induction of apoptosis in A549 cells was investigated using flow cytometric analysis. LPS significantly increased the proportion of A549 apoptotic cells ~2.3-fold compared with the untreated control (Fig. 2A). Furthermore, Hoechst 33258 staining demonstrated increased numbers of apoptotic cells following treatment with LPS (Fig. 2B). ROS generation following LPS treatment was also examined. As presented in Fig. 2C, treatment with LPS significantly increased ROS generation in A549 cells in a time-dependent manner. Western blot analysis demonstrated that LPS treatment markedly decreased the protein level of SIRT1, whereas the acetylated and total p53 protein expression were increased. Additionally, the expression of the pro-apoptotic protein Bax was significantly induced, whereas the expression of Bcl-2 was suppressed (Fig. 2D). These results suggest that LPS treatment induces a pro-apoptotic effect in A549 cells.

An external file that holds a picture, illustration, etc.
Object name is ol-15-06-9609-g01.jpg

LPS induces apoptosis in A549 cells. (A) Flow cytometric analysis. (B) Hoechst 33258 staining. (C) Detection of ROS generation. (D) Western blot analysis. *P<0.05, **P<0.01 vs. the untreated control. LPS, Pseudomonas aeruginosa lipopolysaccharide; DMSO, dimethyl sulfoxide; PI, propidium iodide; Ace, acetylated; ROS, reactive oxygen species; SIRT1, sirtuin 1; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X protein.

LPS induces autophagy in A549 cells 

The effect of LPS treatment on the induction of autophagy was also investigated. Rap was used as a positive control. Electron microscopy demonstrated that LPS induced autophagy in A549 cells in a similar pattern to that induced by Rap (Fig. 3A). Following GFP-LC3 transfection increased numbers of autophagic vesicles (autophagosomes) were observed in A549 cells treated with LPS or Rap (Fig. 3B). Western blot analysis indicated that treatment with LPS significantly decreased p62 protein expression and increased the LC3II/LC3I ratio (Fig. 3C).

An external file that holds a picture, illustration, etc.
Object name is ol-15-06-9609-g02.jpg

LPS induces autophagy in A549 cells. (A) Electron microscopy. (B) Green fluorescent protein-LC3 transfection. (C) Western blotting analysis. *P<0.05 vs. the untreated control. LPS, Pseudomonas aeruginosa lipopolysaccharide; DMSO, dimethyl sulfoxide; Rap, rapamycin; LC3, microtubule-associated protein 1A/1B-light chain 3.

TP inhibits the LPS-induced apoptosis and autophagy in A549 cells by increasing SIRT1 protein expression 

A549 cells were treated with 10 µg/ml TP for 48 h and total protein was extracted from cells. As presented in Fig. 4A, treatment with TP significantly increased the protein expression of SIRT1. ROS generation following LPS treatment with or without 10 µg/ml TP was also examined. TP treatment significantly decreased ROS generation in LPS-treated A549 cells (Fig. 4B). Furthermore, the effect of TP on the induction of apoptosis in A549 cells treated with LPS was also investigated and it was demonstrated that treatment with TP reversed the LPS-induced apoptosis (Fig. 4C). Additionally, western blot analysis demonstrated that transfection with a small interfering RNA targeting SIRT1 inhibited the expression of SIRT1 even in the presence of TP. Furthermore, the expression levels of acetylated and total p53 were also reversed by TP treatment, whereas the protein expression of p62 and Bcl-2 was induced and the protein expression of Bax was suppressed. These results indicate that TP serves an anti-apoptotic role in A549 cells treated with LPS and that this effect is abolished by SIRT1 silencing (Fig. 4D).

 
An external file that holds a picture, illustration, etc.
Object name is ol-15-06-9609-g03.jpg

TP inhibits LPS-induced apoptosis in A549 cells by increasing the protein expression of SIRT1. (A) Treatment with TP increased the protein expression of SIRT1. (B) TP treatment significantly decreased ROS generation in LPS-treated A549 cells. (C) Treatment with TP inhibited the induction of apoptosis in LPS-treated A549 cells. (D) Western blot analysis. *P<0.05, **P<0.01 vs. the untreated control. TP, Tremella polysaccharides; LPS, Pseudomonas aeruginosa lipopolysaccharide; SIRT1, sirtuin 1; Con, control; DMSO, dimethyl sulfoxide; ROS, reactive oxygen species; PI, propidium iodide; Ace, acetylated; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X protein; si, small interfering.

Discussion

P. aeruginosa is associated with hospital-acquired infections frequently in pediatric patients (17,18). LPS is considered a major virulence factor of P. aeruginosa (19,20). LPS infection in pediatric patients may lead to the self-limited upper respiratory tract infection or may develop into severe lower respiratory tract disease often associated with airflow obstruction and high mortality among children. Therefore, the research of therapeutic improvement of this infection is of great importance.

In the present study, it was initially demonstrated that LPS inhibited the viability of A549 cells in a time- and dose-dependent manner. LPS infection appears to exert a deleterious effect on A549 cells, indicating a potential threat of lung injury. The effect of LPS treatment on the induction of apoptosis in A549 cells was also investigated and LPS was demonstrated to induce apoptosis in A549 cells. Previous studies have demonstrated that ROS generation serves a critical role in bacteria-associated cell death (21,22). Therefore, it was hypothesized that LPS-induced apoptosis may be associated with ROS generation. It was demonstrated that LPS increased ROS generation in a time-dependent manner, suggesting that increased ROS production serves a crucial role in the apoptotic cell death induced by P. aeruginosa infection.

It has been demonstrated that SIRT1 serves a key role in several cellular functions, including cell survival, stress resistance and apoptosis (10,23). p53 is a well-characterized SIRT1 substrate (24). It has been reported that SIRT1 deacetylates p53 and decreases the expression of downstream genes, including Bax (24). However, very little is known about the association between SIRT1 and P. aeruginosa infection. The aim of the present study was to evaluate the role of SIRT1 in LPS-induced apoptosis in A549 cells. The results of the present study demonstrated that LPS treatment decreased the protein expression of SIRT1 in A549 cells and indicated that suppressed SIRT1 expression may be associated with LPS-induced lung injury.

Several pharmacological activities, including anti-diabetic and anti-inflammatory action, have been attributed to TP (25–27). However, to the best of our knowledge, the role of TP in the prevention of LPS-induced lung injury has not been investigated to date. The results of the present study demonstrated that treatment with TP inhibited LPS-induced apoptotic cell death in A549 cells. Furthermore, ROS generation induced by LPS was significantly decreased following TP treatment of A549 cells, suggesting a protective role of TP in lung injury. The expression of SIRT1 following TP treatment was also investigated, and it was demonstrated that TP activates SIRT1 expression leading to decreased acetylation of p53, decreased Bax expression and subsequently inhibiting LPS-induced apoptosis.

Autophagy has a dual role in the regulation of cell death. In certain situations, it serves a protective role against harmful conditions promoting cell survival or it may induce programmed cell death, termed autophagic cell death (28,29). Previous studies investigated the association between autophagy and apoptosis (30,31). A number of drugs are known to activate apoptotic and autophagic pathways. For instance, ceramide has been described to induce apoptosis and autophagy in breast and colon cancer cell lines, respectively (32). The antibacterial drug chloroquine chloride has been demonstrated to induce autophagy and apoptotic cell death in leukemia and myeloma cells by inhibiting the mammalian target of rapamycin signaling pathway (33). In the present study, the effect of TP on the induction of autophagy in A549 cells was investigated. It was demonstrated that LPS induced autophagy, which was inhibited by treatment with TP. Notably, this effect could be reversed by knockdown of SIRT1 in A549 cells.

The results of the present study demonstrated that TP induces the expression of SIRT1 and inhibits the LPS-induced ROS generation, autophagy and apoptotic cell death in A549 cells. The results of the present study may have important clinical implications for the treatment of LPS-associated disease in patients with lung injury.