Oxidative stress modulates the expression of toll‑like receptor 3 during respiratory syncytial virus infection in human lung epithelial A549 cells

  • Authors:
    • Min‑Min Wang
    • Min Lu
    • Chuan‑Long Zhang
    • Xuan Wu
    • Jing‑Xian Chen
    • Wei‑Wei Lv
    • Tao Sun
    • Huan Qiu
    • Sheng‑Hai Huang
  • View Affiliations

  • Published online on: May 29, 2018     https://doi.org/10.3892/mmr.2018.9089
  • Pages: 1867-1877
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Toll‑like receptor 3 (TLR3) can react with double stranded RNA and is involved in the inflammatory response to respiratory syncytial virus (RSV) infection. Also, oxidative stress has been reported to be involved in RSV infection. However, the correlation between oxidative stress and TLR3 activation during RSV infection is unclear. Therefore, the present study investigated the association between TLR3 expression and oxidative stress modulation during RSV infection in A549 cells. For comparison, seven treatment groups were established, including RSV‑treated cells, N‑acetyl‑L‑cysteine (NAC)+RSV‑treated cells, oxidant hydrogen peroxide (H2O2)+RSV‑treated cells, normal cell control, inactivated RSV control, NAC control and H2O2 control. The mRNA expression changes of TLR3, interferon regulatory factor‑3 (IRF3), nuclear factor‑κB (NF‑κB) and superoxide dismutase 1 (SOD1) were measured using semi‑quantitative reverse transcription‑polymerase chain reaction, and the protein changes of TLR3 and phospho‑NF‑κB p65 were determined using western blot in A549 cells from the different treatment groups. The present study also evaluated the differences in hydroxyl free radical (·OH), nitric oxide (NO) and total SOD activity in the different treatment groups. The results demonstrated that RSV infection of A549 cells increased the levels of ·OH and NO, while decreasing the activity of total SOD. Pretreatment of A549 cells with H2O2 prior to RSV infection upregulated the mRNA and protein expression of TLR3 and NF‑κB, and downregulated the mRNA expression of IRF3 and SOD1, as well as the total SOD activity. When the infected cells were pretreated with NAC, the mRNA and protein expression of these genes were reversed. These variations in the TLR3‑mediated signaling pathway molecules suggested that oxidative stress may be a key regulator for TLR3 activation during RSV infection. RSV‑induced oxidative stress may potentially activate TLR3 and enhance TLR3‑mediated inflammation. These results may provide better understanding of the RSV‑induced inflammatory and immune pathways, and may also contribute to the drug development and prevention of human RSV diseases.

Introduction

Respiratory syncytial virus (RSV), a single-stranded negative-sense RNA virus (1), not only causes severe lower respiratory tract infections in infants worldwide but also leads to hospitalization of infants (24). As one of the main targets for RSV infection, airway epithelial cells play a very important role in host defense system against RSV infection. Recently, cultures of human respiratory epithelium have been applied to study the mechanisms of RSV infection (5). After RSV infection, airway epithelial cells produce a large number of immune-active molecules, including cytokines, chemokines and reactive oxygen species (ROS) to promote the occurrence of infantile bronchiolitis and pneumonia (6).

Oxidative stress is an invariable feature of human lung epithelial cells, resulting in a large amount of ROS productions that modifies and disrupts cellular biomolecules during immune-inflammatory responses to viral infection (7). This injury is mainly the results of additional ROS produced by further increased oxidative stress (8). It has been reported that RSV could induce the RelA activation mediated by ROS signaling (9). The elevation of ROS and IRF3 signals caused by RSV infection in airway epithelial cells could be blocked by anti-oxidants (10). The formation of ROS produces an imbalance between the antioxidant defenses and oxidant molecules such as hydroxy radicals, hydrogen peroxide, and superoxide anion radicals. ·OH provides strong evidence of increased oxidative stress and is an effective initiator of highly reactive lipid peroxidation (11,12). In addition, nitric oxide (NO) is a key mediator for airway inflammation, which promotes the migration of inflammatory cells to the airway (12,13). ROS and free radicals have been shown to act as cellular signaling molecules participating in various molecular and biochemical processes, including pro-inflammatory mediations such as chemokines and cytokines expressions (14). RSV-induced intracellular ·OH and NO may therefore modulate the expression of pro-inflammatory mediators, and oxidative stress may represent an important mechanism for RSV-induced lung pathogenesis. On this basis, Mastronarde et al (15) proposed that antioxidants might be able to block IL-8 production following RSV infection in vivo. In contrast, the activities of antioxidant enzymes (e.g. SOD, glutathione peroxidase, catalase, and glutathione S-transferase) are very important for cellular defense against RSV infection in A549 cells (6). Our group previously revealed that RSV-intranasally-inoculated mice can react to oxidative stress by increasing the malondialdehyde (MDA), NO and ·OH levels, and reducing SOD and GSH activities in lung tissues. The application of melatonin with anti-oxidant and anti-inflammatory functions reversed the pathophysiology by reducing lung inflammation and ameliorated clinical presentations in RSV-infected mice (16), suggesting that oxidative stress is involved in RSV infection.

Toll-like receptors (TLRs) play a fundamental role in human innate anti-microbial immunity and inflammations by recognizing the conserved pathogen-associated molecular patterns (PAMPs) (17,18). Among them, toll-like receptor 3 (TLR3) and TLR7 are considered as the main mediators of viral-induced signal transductions. TLR3, for example, is able to identify double-strand viral genomic RNA and the replicative intermediates of RSV (19), suggesting that TLR3 plays a role in resisting RSV infection in human respiratory system (20). The reaction of TLR3 with dsRNA activates intracellular signaling and promotes the biosynthesis and secretion of cytokines and other inflammatory mediators. Dou et al (21) reported that RSV induced gene expression of TLR3 and TNF-α both in vitro and in mouse lungs. Once TLR3 is activated, its downstream signaling pathway will lead to the activation of nuclear factor-κB (NF-κB) and interferon regulatory factor-3 (IRF3) (22). NF-κB has been shown to modulate the production of pro-inflammatory cytokines, such as IL-1β, TNF-α and the neutrophil chemoattractant IL-8 (23), which are strongly associated with the outcome of inflammatory disease. Whereas IRF3 was shown to regulate the type I interferon (IFN) expressions (24). Recently, an increase in TLR3 expression was observed in airway epithelial cells of patients with acute respiratory distress syndrome under airway exposure to hyperoxic conditions (25), enhanced TLR3 responses to oxidative stress have also been found in airway epithelial cells (26). These phenomena suggest that oxidative stress may participate in the regulation of TLR3 expression.

N-acetyl-L-cysteine (NAC) is a thiol compound that directly used as a free radical scavenger and a reduced glutathione (GSH) precursor (27), which allow it to be used as an antioxidant in a broad spectrum (28). On the contrary, hydrogen peroxide (H2O2) induces oxidative and inflammatory responses in epithelial cells and it can be used as an oxidant (29).

To our knowledge, by now, there is no study reporting that whether RSV infection increases TLR3 signaling in airway epithelial cells through oxidative stress induction. In order to understand the relationship between TLR3 expression and oxidative stress modulation during RSV infection in A549 cells, we studied the intervening effects of oxidative stress agonist hydrogen peroxide (H2O2) and inhibitor N-acetyl-L-cysteine (NAC) on TLR3 expression. Besides, we proposed that oxidative stress induced by RSV infection might serve as one of the key events in the process of TLR3 activation We hoped that our study would provide a potential new pharmacological method to improve RSV-induced acute lung inflammation.

Materials and methods

Cells, viruses and reagents

The human lung adenocarcinoma alveolar basal epithelial cell line A549 (ATCC® CCL-185®; American Type Culture Collection, Manassas, VA, USA) was gifted by Professor Hai-Ming Wei, Institute of Immunology, University of Science and Technology of China (USTC, Hefei, Anhui, China) and maintained in Dulbecco's modified Eagle's medium (DMEM; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin (100 U/ml)-streptomycin (100 µg/ml) at 5% CO2 and 37°C.

The laryngeal epithelial carcinoma HEp-2 cell line was maintained in our laboratory. Though it has been reported being contaminated by HeLa cells, an epithelium-like cell from a cervical adenocarcinoma (iclac.org/databases/cross-contaminations/), Nevertheless, HEp-2 cells are still a good substrate for RSV (30). Additionally, RSV used in this study was harvested from culture supernatant and was further purified by density gradient centrifugation; therefore, either HEp-2 or HeLa cells would have no intervening effect on the result interpretation of the analysis.

The RSV Long strain was also gifted by Professor Hai-ming Wei, Institute of Immunology, University of Science and Technology of China (USTC, Hefei, Anhui, China) and was multiplicated in HEp-2 cells. Then, the culture supernatant was precipitated by polyethylene glycol 4000, followed by centrifugation on 35–65% discontinuous sucrose gradients. Purified virus suspension was aliquoted, quickly frozen, and stored in liquid nitrogen. The viral titer of purified RSV reached 7×106 PFU/ml as measured by a methylcellulose plaque assay in HEp-2 cells (31).

The UV-inactivated RSV was prepared as follows: A one ml RSV pool was transferred to a culture plate. The plate was placed under a germicidal lamp TUV-15 W/G15 T8 (Philips, The Netherlands) and irradiated at a distance of 10 cm in 3-min intervals, with swirling between the intervals, for a total of 30 min. UV-inactivated virus titers were also determined by plaque assays on HEp-2 cells to confirm the inactivation effect, then the virus stock was stored in liquid nitrogen until use.

H2O2 and NAC were purchased from Sigma Corporation (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). Commercial assay kits for measuring ·OH, NO and SOD activities were purchased from Nanjing Jiancheng Bioengineering Institute, (Nanjing, China).

Experimental design and sample collection

Seven experimental groups were assigned: normal groups (untreated and uninfected with RSV); H2O2 control groups (pretreated with H2O2 without RSV infection); NAC control groups (pretreated with NAC without RSV infection); RSV infection control (cells infected with RSV but no pretreatment); NAC+RSV groups (pretreated with NAC at 5 mM prior to RSV infection), H2O2 +RSV groups (pretreated with H2O2 at 150 µM prior to RSV infection) and inactivated RSV groups (cells infected with inactivated RSV but no pretreatment). The pretreatment time with H2O2 or NAC for the corresponding group was 1 h. Cells in the corresponding groups were infected with RSV at MOI=1 in serum-free mediums for 2 h before fresh mediums were added. The culture supernatants and trypsinized cells were separately collected by centrifugation at 4, 8, 12 and 24 h post infection (pi) and the samples were assessed by the following experiments.

Measurement of ·OH and NO in culture supernatants

·OH concentration was determined using a commercial ·OH assay kit based on the Fenton reaction method according to the manufacturer's protocol (Nanjing Jiancheng Bioengineering Institute) (32). Briefly, the reaction mixtures were added to a quartz capillary tube, then Griess chromogenic reagent was added to form a colored substance for another 20 min at room temperature (RT) (16). The color depth of the substance is proportional to the amount of ·OH (33). The absorbance values at 550 nm were recorded, and the data was expressed as units/ml.

NO concentration was determined using a commercial NO assay kit based on the Griess reaction method according to the manufacturer's instructions (Nanjing Jiancheng Bioengineering Institute) (34). First, 50 µl of Griess reagent I was added to 100 µl of the supernatant from A549 cultures. Second, 50 µl of Griess reagent II was added. Third, the reaction mixture was incubated for 10 min at RT, and 160 µl of the supernatant was used for detection after centrifugation at 3500 rpm for 15 min. Last, the absorbance at 550 nm was recorded, and the results were expressed as µmol/l. All samples were tested in triplicate.

Measurement of total SOD activity in A549 cells

Proteins from A549 cells were prepared as a method previously described (16). First, the cells were washed with PBS before the treatment with Versene-Trypsin PBS solution. Then, the trypsin-digested cells were centrifuged at 1,400 g for 10 min and the pellet was re-suspended in a lysis buffer supplemented with protease inhibitors. Next, after 30 min of incubation on the ice, the extraction mixture was centrifuged at 12,000 g at 4°C for 30 min. Last, the supernatant was transferred to a fresh tube, and its protein concentration was measured by the Lowry method.

Superoxide dismutase (SOD), an important enzyme in the antioxidant system, is able to convert O2 to hydrogen peroxide (H2O2) and eventually change them to water. The mechanism for total SOD assay was based on its ability to inhibit the oxyamine oxidation within the xanthine/xanthine oxidase system (35). SOD activity was determined using a commercial WST-1 assay kit (Nanjing Jiancheng Bioengineering Institute). The absorbances at 450 nm at the reaction endpoint were read by a microplate reader (ELX800UV; BioTek Instruments, Inc., Winooski, VT, USA). The SOD activity of each sample was calculated using a previously described equation (36). All samples were tested for three times, and the results were expressed as units per mg protein.

TLR3, NF-κB p65, IRF3 and superoxide dismutase 1 (SOD1) mRNA semi-quantification by reverse transcription polymerase chain reaction (RT-PCR). The kinetics of gene expression of TLR3, NF-κB p65, IRF3 and SOD1 were analyzed by semi-quantitative RT-PCR. Total RNA was extracted from A549 cells with TRI Reagent™ (Sigma-Aldrich; Merck KGaA) following the manufacturer's instructions. Then, the total RNA was treated with RNase-free DNase I (Invitrogen; Thermo Fisher Scientific, Inc.) to remove genomic DNA contamination and reverse-transcribed into cDNA using Thermo Scientific Revert Aid First Strand cDNA Synthesis kit (Fermentas; Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. Next, the synthesized first strand cDNA was amplified by PCR with the primers for TLR3, NF-κB p65, IRF3, or SOD1 genes. The PCR primers (Table I) for TLR3, NF-κB p65, IRF3, SOD1 and β-actin genes were designed using the Primer Express software, as previously described (37). The reaction conditions and cycle numbers used for the PCR of each gene were shown in Table II. PCR products and a DNA molecular weight marker (DL2000; Takara Biotechnology Co., Ltd., Dalian, China) were electrophoresed in a 1.5% agarose gel and visualized under UV light. The density of the bands was quantified by densitometry using Labworks software, and the expression levels were expressed as the fold-increase compared to β-actin controls.

Table I.

Primer sequence and length for polymerase chain reaction products of each human gene.

Table I.

Primer sequence and length for polymerase chain reaction products of each human gene.

GeneForward primer (5′-3′)Reverse primer (5′-3′)Size (bp)
TLR3 GATCTGTCTCATAATGGCTTG GACAGATTCCGAATGCTTGTG304
NF-κB p65 CACAAGGAGACATGAAACAG CCAGAGACCTCATAGTTGT187
SOD1 ATGGCGACGAAGGCC TTATTGGGCGATCCC465
IRF3 GACCTCACGACCCACATAA ACCCCACCAGCCGCAGGCCC377
β-actin GGCTTTGAGTAATGAGAATTTCGA ATCAGTTGCAATCAAGAAGTGTTG520

[i] TLR4, Toll-like receptor 3; NF-κB p65, nuclear factor-κB p65; SOD1, superoxide dismutase 1; IRF3, interferon regulatory factor-3.

Table II.

Polymerase chain reaction thermocycling conditions and cycle numbers for each gene.

Table II.

Polymerase chain reaction thermocycling conditions and cycle numbers for each gene.

GeneInitial DenatureDenatureAnnealingExtensionNo. of cycles
TLR394°C for 5 min94°C for 40 sec55°C for 45 sec72°C for 1 min32
NF-κB p6594°C for 5 min94°C for 40 sec57.7°C for 45 sec72°C for 1 min32
SOD194°C for 5 min94°C for 40 sec57°C for 45 sec72°C for 1 min32
IRF394°C for 5 min94°C for 40 sec49.4°C for 45 sec72°C for 1 min32
β-actin94°C for 5 min94°C for 40 sec55°C for 45 sec72°C for 1 min32

[i] TLR4, Toll-like receptor 3; NF-κB p65, nuclear factor-κB p65; SOD1, superoxide dismutase 1; IRF3, interferon regulatory factor-3.

TLR3 and p-NF-κB protein quantification by western blot

The protein level changes of TLR3 and p-NF-κB were analyzed by western blot assay. First, the cells in each group were washed with PBS solution and lysed in a lysis buffer containing phenylmethylsulfonyl fluoride (PMSF). All samples were incubated on ice for 30 min and centrifuged at 11,000 g for 5 min. Second, the protein concentrations in each group of centrifuged supernatant were determined by the Lowry method. Third, fifty µg of protein was run on a 10% SDS-PAGE and then transferred to a polyvinylidene fluoride (PVDF) membrane (sc-296042; Santa Cruz Biotechnology, Inc., Dallas, TX, USA). Fourth, nonspecific binding sites were blocked with 5% nonfat milk in Tris-buffered saline with Tween 20 (TBST) for 2 h at RT. Fifth, the membranes were incubated with rabbit anti-TLR3 antibody (sc-28999; Santa Cruz Biotechnology Inc, Santa Cruz, CA, USA) and anti-p-NF-κB antibody (sc-33020; Santa Cruz Biotechnology Inc.) separately at a 1:1,000 dilution overnight at 4°C. After washing with TBST for 3×10 min, the membranes were incubated with a horseradish peroxidase-conjugated anti-rabbit IgG (1:10,000) (sc-2004; Santa Cruz Biotechnology Inc.) for 2 h at RT. Last, the blots were washed, and the antigens were visualized using an enhanced chemiluminescence western blot detection system (SuperSignal West Femto kit; Thermo Scientific, Inc.) and recorded by a Tanon 4500 automatic digital gel image analysis system (Tanon 4500; Tanon, Shanghai, China).

A β-actin monoclonal antibody (1:1,000; TA-09; OriGene Technologies, Inc., Beijing, China) and horseradish peroxidase-conjugated sheep anti-mouse IgG (1:10,000; sc-2005; Santa Cruz Biotechnology Inc.) were used as the internal control, The molecular weights of TLR3, NF-κB and β-actin in the PAGE gel were 104, 65 and 43 KDa, respectively. The density of the bands on the blot was quantified using Labworks imaging software. Compared with β-actin control group, densitometry in each group was expressed as the fold-increase.

Statistical analysis

The resulting data are recounted as the mean ± standard error mean. One-way analysis of variance analysis and the Fisher post hoc test were performed to determine statistical significance among each group using SPSS v.17.0 software (SPSS Inc., Chicago, IL, USA). P<0.05 for the null hypothesis was considered to indicate a statistically significant difference.

Results

The releasements of ·OH and NO after different treatments

Oxidative stress can be induced from either excess ROS generation or impaired antioxidant capacity, which produced free radicals and reactive oxygen molecules in activated cells (11). Because that oxidative stress-mediated events were correlated with the releasement of ·OH and NO, therefore, ·OH generation was considered as a marker of free oxygen species due to the extreme instability of ROS. In this study, the NO and ·OH in A549 cells from inactivated-RSV control, NAC control and H2O2 control groups, at different time points, had no significant changes compared to the normal control group within 24 h (Fig. 1). However, in both the RSV-infected group and the H2O2+RSV-treated group (Fig. 1A), the ·OH concentration increased dramatically compared to the normal cell control group (P<0.01) Nevertheless, in cell group treated with NAC+RSV (Fig. 1A), although the ·OH levels also increased with an increasing time of RSV infection, they were significantly lower than those in the RSV-infected group at the corresponding time points. Thus, the ·OH elevations were associated with the extended time duration of RSV infection, suggesting ·OH in A549 cells was up-regulated in a time-dependent manner after RSV infection. On the results of NO concentration, its variation had a similar change pattern to that of ·OH concentration (Fig. 1B), which also demonstrated that the RSV infection induced a significant elevation of oxidative stress in A549 cells. Notably, UV-inactivated RSV did not elevate the ·OH and NO levels compared with the normal group (data not shown), suggesting that the alteration of oxidative stress is dependent on virus replication.

The total SOD activity after different treatments

The protein level and activity of SOD are important indicators for the cellular antioxidant stress capacity, and increased SOD expression can contribute to antioxidant functions in vivo (16). As shown in Fig. 1C, the SOD levels were significantly decreased in a time-dependent manner in the RSV-infected group compared with those in the normal group at all time points tested (P<0.01; Fig. 1C). In contrast, although pretreatment of A549 cells with inactivated-RSV or NAC or H2O2 alone had no impact on SOD release within 24 h, the administration of NAC+RSV, but not H2O2+RSV, was able to markedly improve SOD activity during infection compared with RSV-infected group (Fig. 1C). Thus, the antioxidant capacity of RSV-infected A549 cells was enhanced by the pretreatment of NAC, which suggested that oxidative stress damage indeed occurred after RSV infection.

The mRNA levels of TLR3, NF-κB p65, IRF3 and SOD1 genes after different treatments

TLR3 activation and the mRNA expression levels of its downstream molecules, including NF-κB p65, IRF3 and SOD1, were analyzed by semi-quantitative RT-PCR in this study. Figs. 2 and 3 showed the mRNA expression of TLR3, NF-κB p65, SOD1 and IRF3 in A549 cells with RSV infection (Figs. 2A and 3A) and cultures pretreatment of H2O2 (Figs. 2D and 3B) or NAC (Figs. 2E and 3C). The results showed that adding inactivated-RSV (data not shown) or H2O2 (Fig. 2B) or NAC (Fig. 2C) alone has no effect on TLR3 mRNA expression in A549 cells. However, the RSV infection treatment and H2O2+RSV treatment significantly elevated both TLR3 (Fig. 2F) and NF-κB p65 (Fig. 3D) mRNA levels at all time points tested. Conversely, the mRNA expression of TLR3 and NF-κB was decreased in the NAC+RSV group compared to the RSV group, which suggested that NAC played an inhibitory role in RSV-induced TLR3 and NF-κB activation.

In RSV-treated and RSV+H2O2-treated groups, the levels of IRF3 and SOD1 mRNA were markedly decreased with statistically significant differences, compared to the normal cell group (Fig. 3). This reduction could be reversed by the NAC+RSV treatment, but the mRNA levels of IRF3 and SOD1 in NAC+RSV-treated group were still less than that shown in the RSV-treated group.

Therefore, the RSV infection in A549 cells enhanced the mRNA expression of TLR3 and NF-κB p65 but suppress the mRNA expression of IRF3 and SOD1, indicating that the oxidative stress inhibitor and agonist altered TLR3 activation and the expression of relative downstream signaling molecules at the transcriptional level. These data are in line with the results described above with the determination of ·OH and NO concentration and total SOD activity.

The protein expression of TLR3 and p-NF-κB after different treatments

Because that TLR3 was recently reported to be able to recognize the viral dsRNA intermediates produced during RSV replication and activate NF-κB (20,22), the expression of both TLR3 and p-NF-κB proteins were studied by western blot assay. It was found that there were no significant differences in TLR3 and p-NF-κB changes in inactivated-RSV control or H2O2 control or NAC control groups at the indicated concentrations (Fig. 4), therefore the use of inactivated-RSV or H2O2 or NAC alone had no effect on the experimental results. As shown in Fig. 4, TLR3 and p-NF-κB protein were significantly increased in the RSV-treated and the H2O2+RSV-treated groups compared to the normal cell control group. However, the treatment with NAC+RSV attenuated the RSV-induced elevation of TLR3 and p-NF-κB protein, suggesting that NAC may inhibit RSV-induced TLR3 activation (Fig. 4F).

These results demonstrate that the elevated TLR3 expression is consistent with the increasing of p-NF-κB protein expression during RSV infection, and the TLR3 activation can enhance the up-regulation of its downstream signaling proteins, including NF-κB.

Discussion

RSV is a highly pathogenic virus that can lead to severe respiratory diseases in newborns, children, the elderly and individuals with immune impairment (38,39). It has been reported that airway inflammation plays a crucial role in the disease outcome in RSV-infected hosts (40). Although the pathogenesis of RSV infection remains largely unknown, previous studies have suggested that a relative overload of oxidants in response to RSV infection in human airway epithelial cells may have an important impact on lung injury (41).

Oxidative stress was reported to induce the production of reactive oxygen species (ROS), causing oxidative damage in tissue during RSV infection in vivo (11,42). Also, the decrease of SOD total activity can result in an excess availability of superoxide and generate hydroxyl radicals that are associated with the initiation and propagation of lipid peroxidation (43). Though a study has reported that RSV infection of A549 cells was able to induce a significant decrease in SOD1, SOD3, GST and catalase gene expressions along with the increase of SOD2; however, the total SOD activity would decrease initially, followed by a subsequent increase after 24 h (41). The latter observations are consistent with our findings in the current study, namely that total SOD activity is decreased within 24 h of RSV infection.

The activation of TLR2, TLR3, TLR4, and TLR7/8 in the innate immune system can lead to the strong up-regulation of SOD2 gene expression in macrophages during microbial infection (44). To et al (45) had demonstrated that influenza A virus-induced TLR7 activation enhanced the oxidative burst of NOX2-oxidase dependence in macrophages, suggesting the occurring of acute lung injury after influenza A virus infections. Although oxidative stress was recently found to augment the response of TLR3 to dsRNA in airway epithelial cells through NF-κB pathway (26), to date, there is no study described the relationship between RSV infection with both TLR3 activation and oxidative stress generation, therefore, we performed the experiments and firstly reported that the elevated expression of TLR3 could be modulated by oxidative stress during RSV infection.

TLR3 gene expression may be regulated by RIG-I-induced IFN-β secretion in the early response of host cells to RSV infection and TLR3 indeed mediates epithelial responses to RSV infection (46). Our group previously found that RSV infection induced TLR gene transcription by recognizing the viral dsRNA genome, and it is likely that RSV infection promotes a rapid activation of innate responses via the increased expression of TLR3 (47). However, the molecular mechanism for RSV-induced TLR3 up-regulation was unclear (48,49). In the present study, RSV infection was shown to up-regulate both mRNA and protein expression levels of TLR3 in A549 cells, furthermore, it was also shown that RSV-induced, TLR3-mediated early signal events could lead to the activation of NF-κB and IRF3, both of which were two key transcriptional factors for the expression of inflammatory cytokines and chemokines in airway epithelial cells. Pretreatment with H2O2 before RSV infection increases TLR3 expression and TLR3-mediated NF-κB activity, whereas pretreatment with antioxidant NAC inhibits the activation of TLR3 pathways, including pNF-κB expression (Figs. 24). These results are in agreement with our previous finding that RSV can induce TLR3 expression and NF-κB/RelA subunit phosphorylation and transcriptional activation (30).

Matsukura et al (50) reported that dsRNAs such as poly (I:C) bound to TLR3 that distributed on the cell surface and induced some chemokines and cytokines gene expressions through activation of NF-κB and IRF3. Their results indicated that dsRNA could increase the expression of inflammatory cytokines and chemokines via TLR3-NF-κB and TLR3-IRF3 signaling in airway epithelial cells (50,51). Liu et al (46) showed that the TLR3 knockdown mediated by siRNA significantly reduced NF-κB/RelA transcriptional levels by blocking the activating phosphorylation of NF-κB/RelA at serine residue 276. It was also demonstrated that NADPH oxidase 2 (NOX2) mediates ROS production in RSV-infected human airway epithelial cells and that NOX2 acts upstream of the phosphorylation of both IκBα at Ser32 and of p65 at Ser536 in RSV-infected A549 cells and in human bronchial epithelial cells (52). Jamaluddin et al (9) demonstrated the RSV-induced ROS induced activation of RelA and RSV-induced ROS formation also resulted in STAT activation and IRF gene expression induction (10). Unanimously, our current study revealed that oxidative stress was involved in RSV-augmented NF-κB and IRF activation in airway epithelial cells and lead to an increase of TLR3 expression.

It is worth noting that preferential induction of TLR3 is also observed in human astrocytes after its exposure to H2O2 to induce oxidative stress (53) and that H2O2, but not poly (I:C), appears to activate NF-κB and nuclear translocation of p65 in human SH-SY5Y neuroblastoma cells (54). ROS may potentiate TLR3 expression through NOX2 signaling by RSV infection (55).

As far as the investigations are concerned, we further confirmed that there were no significant differences in TLR3 signaling in A549 cells pretreated with either H2O2 or NAC without RSV infection (Fig. 2B, C and Fig. 4A, B), as previously described (26,56). Geiler et al (56) reported that there was no significant decrease in virus titer produced in A549 cells as a result of NAC treatment (5 mM) at 48 h pi with influenza A virus; thus, we chose to use NAC at 5 mM in the current study. Unfortunately, this concentration of NAC did not fully recover SOD activity to the normal levels, nor did we detect the effect of H2O2 on virus titer, both of which are the limitations of the present study.

The transcriptional activators of the NF-κB family participated in the modulation of cell proliferation, differentiation, apoptosis, inflammation, immunity and cytokine expression in response to various types of stimulation (57). In the inactive state, the NF-κB is mainly located in the cytoplasm, forming complexes with IκB. Once it is stimulated by extracellular stimuli such as viruses, IκB can be rapidly phosphorylated and degraded, allowing the release of NF-κB p65 and subsequent NF-κB p65 translocation to the nucleus with the increase of NF-κB regulated gene expression (58). IRF3 acts as an important transcriptional regulator in antiviral immune responses, viral infection can also induce the IRF3 phosphorylation and its translocation to the nucleus (50). The binding of IRF3 to IFN-stimulated response element (ISRE) in the promoters of type I IFN genes is thought to activate the transcription of these genes (59). However, non-structural proteins (NS1 and NS2) of RSV have been demonstrated to inhibit the induction of IFN-α/β in A549 cells and human macrophages (60,61). It was proposed that RSV NS1 protein limits IRF3 nuclear translocation through blocking its phosphorylation by decreasing the levels of 2 kinases upstream of IRF3 activation, TRAF3 and IKKε (62). RSV NS2 protein was also proposed to act on downstream molecules such as TRAF3 and ultimately inhibit IFN-β expression. Our results found that RSV infection indeed inhibits early IRF3 activation at 24 h pi, which is consistent with Hosakote's study (63). Moreover, oxidant H2O2 pretreatment further inhibits the early activation of IRF3 (Fig. 3F), indicating that an RSV-mediated redox-sensitive pathway probably inhibits IRF3 activation. Further studies can be done to investigate the mechanisms of interaction between NF-κB and IRF under conditions of RSV infection.

This study was conducted using A549 cells derived from human lung cancer tissue, which is similar to previous studies (41,56,6466). A549 cells have also been widely used in the research of oxidative stress by RSV infection (41,64,67) or the alteration of TLR3 expression by RSV (21,68). Therefore, we believe that A549 cells, as an adenocarcinomic alveolar basal epithelial cell line, will have no intervening effect on the analysis and conclusion of the study. However, it will be interesting to investigate whether H2O2 and NAC have the same effects on RSV-induced TLR3, NF-κB and IRF3 expression in non-carcinomic primary epithelial cells in the future.

For production of type I IFN, RSV is generally considered as a poor inducer of IFN-α and IFN-β, comparing with other RNA viruses (69,70). However, RSV is previously reported to induce high levels of IFN-β expression in cultures of various types of human fibroblasts, respiratory epithelial cells, and mesenchymal stem cells (MSCs) (7173). Besides, RSV is also reported to induce high levels of IFN-α expression in different subsets of dendritic cells (DC) (7477). Furthermore, RSV treatment is also reported to induce a type I IFN response in both human cord blood-derived mast cell (CBMCs) and peripheral blood derived mast cells (78,79). Therefore, the results on the type I IFN production in responding to RSV infection are controversial and it seems that many different factors can determine the type I IFN production under the infection of RSV, such as different cell types and different viral strains (71,72,7477,80). On these basis, we tentatively thought that the response of IFN-α and IFN-β to the infection of RSV long strain in A549 cells, which was related to the experiments in this study, might have various possible results. Thus, for the reason that the inducement of type I IFN in A549 cells may not be representative, we consider that it is not necessary to detect IFN-α and IFN-β in this study.

Taken together, we investigated the relationship between TLR3 expression and oxidative stress modulation in RSV-infected A549 cells in this study. We used oxidative stress agonist H2O2 and inhibitor NAC (equivalent to oxidant and antioxidant) to interfere with RSV infection from both positive and negative sides to determine the effect of oxidative stress on TLR3 expression and TLR3-mediated inflammatory and immune pathways. Our results showed that, in RSV-infected A549 cells, the production of hydroxyl free radical (·OH) and nitric oxide (NO) were induced, while the superoxide dismutase (SOD) activity was reduced. On the variation of gene expression, our results showed that both of mRNA and protein expression levels of TLR3 and NF-κB were up-regulated. Pretreatment of H2O2 plus RSV infection enhanced RSV-induced TLR3 and NF-κB expression, whereas Pretreatment of NAC plus RSV infection reduced them. These results indicated that oxidative stress was a critical regulator of TLR3 activation in RSV infection and suggested that oxidative stress might potentiate increasing the TLR3 expression in A549 cells after RSV infection, which might partly explain the enhancement of the associated downstream signaling pathway, including NF-κB activity. The elevated TLR3 and NF-κB activities might be key factors in interpreting oxidative stress effects induction. These findings may contribute to the study of RSV pathogenesis and the development of RSV prevention and control.

To further confirm the role of oxidative stress discovered in this study, we plan to knock-out TLR3 and p-NF-κB genes using the CRISPR/Cas9 technique in RSV future work. First, we will synthesize three high-grade small-guide RNAs (sgRNAs) that could specifically identify TLR3 and p-NF-κB genes and inserted them into lenti CRISPRv2 plasmid. Second, 293T cells will be transfected with the recombinant sgRNA-lenti CRISPRv2 plasmid to yield subsequent sgRNA-Cas9 lentivirus prior to its further infection of A549 cells. Third, positive A549 cells will be screened using puromycin and be validated by PCR and western blot. Last, sequencing analysis will be adopted to confirm the mutation site of the obtained genes-knockout A549 cells.

Besides, we also plan to over-express TLR3 and p-NF-κB genes by lentiviral transfection to verify the function of oxidative stress. First, we will amplify TLR3 and p-NF-κB genes by RT-PCR and insert them into the lentiviral vector pLENTI-cGFP using the homologous recombination method. Second, the constructed recombinant vector will be confirmed by DNA sequencing and be co-transfected with psPAX2 and pMD2. G helper plasmids into HEK293T packaging cells to produce the lentiviral particles on the ratio of 3:2:1 by PEI transfection reagent. Third, the lentiviral particles will be transduced into A549 cells and the infection efficiency will be measured by Fluorescence Microscopy. Last, the GFP-tag-expressed cells will be sorted by the Flow Cytometer (FCM) after 2 weeks and the protein expressions of TLR3 and p-NF-κB and GFP in the stable cell lines will be confirmed by western blot.

Once the TLR3 and p-NF-κB gene deletion and over-expression are achieved, the established cell lines will be subjected to molecular biological tests in the cases of RSV infection and H2O2 or NAC intervention. We hope that these experiments will facilitate further investigation of the molecular mechanism of oxidative stress modulation of the TLR3 expression in RSV infection.

Acknowledgements

The authors would like to thank Professor Hai-ming Wei (University of Science and Technology of China, Hefei, Anhui, China) for kindly providing the A549 cells and RSV Long strain. The authors are also grateful to Ms. Xiao-yan Zhang (Department of Microbiology, Anhui Medical University, Hefei, Anhui, China) for her technical assistance, and Mr. Hai-yang Yu (Department of Microbiology, Anhui Medical University) for his helpful discussions and suggestions during the preparation of this manuscript.

Funding

The present study was supported by grants from the Natural Science Foundation of China (grant no. 81371797), the Natural Science Foundation of Anhui Province of China (grant no. 1308085MH129) and the Key Project of Natural Science Research of Anhui Education Department (grant no. KJ2012A152).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

MMW and SHH conceived and planned the study. XW performed the statistical analysis and JXC analyzed the data. WWL purified the RSV Long strain, and TS and HQ performed RT-PCR and western blot experiments. MMW, ML and CLZ carried out the chemical detection experiments. MMW, ML and CLZ wrote the manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

dsRNA

double stranded RNA

GSH

glutathione

H2O2

hydrogen peroxide

IFN

interferon

IRF3

interferon regulatory factor-3

ISRE

IFN-stimulated response element

MDA

malondialdehyde

NAC

N-acetyl-L-cysteine

NF-κB

nuclear factor-κB

NO

nitric oxide

NS

nonstructural protein

·OH

hydroxyl free radical

PAMPs

pathogen-associated molecular patterns

PMSF

phenylmethylsulfonyl fluoride

PVDF

polyvinylidene fluoride

ROS

reactive oxygen species

RT-PCR

reverse transcription polymerase chain reaction

RSV

respiratory syncytial virus

SOD

superoxide dismutase

TLRs

Toll-like receptors

TLR3

Toll-like receptors 3

pi

post infection

RT

room temperature

References

1 

Nair H, Verma VR, Theodoratou E, Zgaga L, Huda T, Simões EA, Wright PF, Rudan I and Campbell H: An evaluation of the emerging interventions against Respiratory Syncytial Virus (RSV)-associated acute lower respiratory infections in children. BMC Public Health. 11 Suppl 3:S302011. View Article : Google Scholar : PubMed/NCBI

2 

Piedimonte G: Respiratory syncytial virus and asthma: Speed-dating or long-term relationship? Curr Opin Pediatr. 25:344–349. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Mejías A, Chávez-Bueno S, Gómez AM, Somers C, Estripeaut D, Torres JP, Jafri HS and Ramilo O: Respiratory syncytial virus persistence: Evidence in the mouse model. Pediatr Infect Dis J. 27 10 Suppl:S60–S62. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Wedzicha JA: Role of viruses in exacerbations of chronic obstructive pulmonary disease. Proc Am Thorac Soc. 1:115–120. 2004. View Article : Google Scholar : PubMed/NCBI

5 

McCutcheon KM, Jordan R, Mawhorter ME, Noton SL, Powers JG, Fearns R, Cihlar T and Perron M: The interferon type I/III response to respiratory syncytial virus infection in airway epithelial cells can be attenuated or amplified by antiviral treatment. J Virol. 90:1705–1717. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Hosakote YM, Komaravelli N, Mautemps N, Liu T, Garofalo RP and Casola A: Antioxidant mimetics modulate oxidative stress and cellular signaling in airway epithelial cells infected with respiratory syncytial virus. Am J Physiol Lung Cell Mol Physiol. 303:L991–L1000. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Bakunina N, Pariante CM and Zunszain PA: Immune mechanisms linked to depression via oxidative stress and neuroprogression. Immunology. 10–Jan;2015.(Epub Ahead of Print). View Article : Google Scholar : PubMed/NCBI

8 

Antonio AM and Druse MJ: Antioxidants prevent ethanol-associated apoptosis in fetal rhombencephalic neurons. Brain Res. 1204:16–23. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Jamaluddin M, Tian B, Boldogh I, Garofalo RP and Brasier AR: Respiratory syncytial virus infection induces a reactive oxygen species-MSK1-phospho-Ser-276 RelA pathway required for cytokine expression. J Virol. 83:10605–10615. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Liu T, Castro S, Brasier AR, Jamaluddin M, Garofalo RP and Casola A: Reactive oxygen species mediate virus-induced STAT activation: Role of tyrosine phosphatases. J Biol Chem. 279:2461–2469. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Miller AL, Bowlin TL and Lukacs NW: Respiratory syncytial virus-induced chemokine production: Linking viral replication to chemokine production in vitro and in vivo. J Infect Dis. 189:1419–1430. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Reiter RJ, Tan DX, Terron MP, Flores LJ and Czarnocki Z: Melatonin and its metabolites: New findings regarding their production and their radical scavenging actions. Acta Biochim Pol. 54:1–9. 2007.PubMed/NCBI

13 

Stark JM, Khan AM, Chiappetta CL, Xue H, Alcorn JL and Colasurdo GN: Immune and functional role of nitric oxide in a mouse model of respiratory syncytial virus infection. J Infect Dis. 191:387–395. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Chtourou Y, Aouey B, Kebieche M and Fetoui H: Protective role of naringin against cisplatin induced oxidative stress, inflammatory response and apoptosis in rat striatum via suppressing ROS-mediated NF-κB and P53 signaling pathways. Chem Biol Interact. 239:76–86. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Mastronarde JG, Monick MM and Hunninghake GW: Oxidant tone regulates IL-8 production in epithelium infected with respiratory syncytial virus. Am J Respir Cell Mol Biol. 13:237–244. 1995. View Article : Google Scholar : PubMed/NCBI

16 

Huang SH, Cao XJ, Liu W, Shi XY and Wei W: Inhibitory effect of melatonin on lung oxidative stress induced by respiratory syncytial virus infection in mice. J Pineal Res. 48:109–116. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Marshak-Rothstein A and Rifkin IR: Immunologically active autoantigens: The role of toll-like receptors in the development of chronic inflammatory disease. Annu Rev Immunol. 25:419–441. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Meylan E and Tschopp J: Toll-like receptors and RNA helicases: Two parallel ways to trigger antiviral responses. Mol Cell. 22:561–569. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Tatematsu M, Nishikawa F, Seya T and Matsumoto M: Toll-like receptor 3 recognizes incomplete stem structures in single-stranded viral RNA. Nat Commun. 4:18332013. View Article : Google Scholar : PubMed/NCBI

20 

Alexopoulou L, Holt AC, Medzhitov R and Flavell RA: Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature. 413:732–738. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Dou Y, Zhao Y, Zhang ZY, Mao HW, Tu WW and Zhao XD: Respiratory syncytial virus infection induces higher Toll-like receptor-3 expression and TNF-α production than human metapneumovirus infection. PLoS One. 8:e734882013. View Article : Google Scholar : PubMed/NCBI

22 

Akira S, Uematsu S and Takeuchi O: Pathogen recognition and innate immunity. Cell. 124:783–801. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Rot A and von Andrian UH: Chemokines in innate and adaptive host defense: Basic chemokinese grammar for immune cells. Annu Rev Immunol. 22:891–928. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Majumder S, Zhou LZ, Chaturvedi P, Babcock G, Aras S and Ransohoff RM: p48/STAT-1alpha-containing complexes play a predominant role in induction of IFN-gamma-inducible protein, 10 kDa (IP-10) by IFN-gamma alone or in synergy with TNF-alpha. J Immunol. 161:4736–4744. 1998.PubMed/NCBI

25 

Murray LA, Knight DA, McAlonan L, Argentieri R, Joshi A, Shaheen F, Cunningham M, Alexopolou L, Flavell RA, Sarisky RT and Hogaboam CM: Deleterious role of TLR3 during hyperoxia-induced acute lung injury. Am J Respir Crit Care Med. 178:1227–1237. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Koarai A, Sugiura H, Yanagisawa S, Ichikawa T, Minakata Y, Matsunaga K, Hirano T, Akamatsu K and Ichinose M: Oxidative stress enhances toll-like receptor 3 response to double-stranded RNA in airway epithelial cells. Am J Respir Cell Mol Biol. 42:651–660. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Cotgreave IA: N-acetylcysteine: Pharmacological considerations and experimental and clinical applications. Adv Pharmacol. 38:205–227. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Jaspers I, Ciencewicki JM, Zhang W, Brighton LE, Carson JL, Beck MA and Madden MC: Diesel exhaust enhances influenza virus infections in respiratory epithelial cells. Toxicol Sci. 85:990–1002. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Song JJ, Lim HW, Kim K, Kim KM, Cho S and Chae SW: Effect of caffeic acid phenethyl ester (CAPE) on H2O2 induced oxidative and inflammatory responses in human middle ear epithelial cells. Int J Pediatr Otorhinolaryngol. 76:675–679. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Collins PL, Chanock RM and Murphy BR: Respiratory syncytial virus, in fields virology. 4th Edition. Knipe D and Howley P: Lippincott Williams & Wilkins; Philadelphia: pp. 1443–1485. 2001

31 

Huang SH, Cao XJ and Wei W: Melatonin decreases TLR3-mediated inflammatory factor expression via inhibition of NF-kappa B activation in respiratory syncytial virus-infected RAW264.7 macrophages. J Pineal Res. 45:93–100. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Lloyd RV, Hanna PM and Mason RP: The origin of the hydroxyl radical oxygen in the Fenton reaction. Free Radic Biol Med. 22:885–888. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Wink DA, Wink CB, Nims RW and Ford PC: Oxidizing intermediates generated in the Fenton reagent: Kinetic arguments against the intermediacy of the hydroxyl radical. Environ Health Perspect. 102 Suppl 3:S11–S15. 1994. View Article : Google Scholar

34 

Kalaivani P, Saranya S, Poornima P, Prabhakaran R, Dallemer F, Padma Vijaya V and Natarajan K: Biological evaluation of new nickel(II) metallates: Synthesis, DNA/protein binding and mitochondrial mediated apoptosis in human lung cancer cells (A549) via ROS hypergeneration and depletion of cellular antioxidant pool. Eur J Med Chem. 82:584–599. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Wang H, Wei W, Shen YX, Dong C, Zhang LL, Wang NP, Yue L and Xu SY: Protective effect of melatonin against liver injury in mice induced by Bacillus Calmette-Guerin plus lipopolysaccharide. World J Gastroenterol. 10:2690–2696. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Zhou JY and Prognon P: Raw material enzymatic activity determination: A specific case for validation and comparison of analytical methods-the example of superoxide dismutase (SOD). J Pharm Biomed Anal. 40:1143–1148. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Balenger SL, McClure CJ and Hill GE: Primer design and transcript quantification of a highly multiplexed RT-PCR for a nonmodel avian species. Mol Ecol Resour. 12:116–122. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Huo X, Fang B, Liu L, Yu H, Chen H, Zheng J, Zhang Y, Xu Z, Klena JD, Varma JK, et al: Clinical and epidemiologic characteristics of respiratory syncytial virus infection among children aged <5 years, Jingzhou City, China, 2011. J Infect Dis. 208 Suppl 3:S184–S188. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Collins PL and Graham BS: Viral and host factors in human respiratory syncytial virus pathogenesis. J Virol. 82:2040–2055. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Segovia J, Sabbah A, Mgbemena V, Tsai SY, Chang TH, Berton MT, Morris IR, Allen IC, Ting JP and Bose S: TLR2/MyD88/NF-κB pathway, reactive oxygen species, potassium efflux activates NLRP3/ASC inflammasome during respiratory syncytial virus infection. PLoS One. 7:e296952012. View Article : Google Scholar : PubMed/NCBI

41 

Hosakote YM, Liu T, Castro SM, Garofalo RP and Casola A: Respiratory syncytial virus induces oxidative stress by modulating antioxidant enzymes. Am J Respir Cell Mol Biol. 41:348–357. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Roy J, Pallepati P, Bettaieb A, Tanel A and Averill-Bates DA: Acrolein induces a cellular stress response and triggers mitochondrial apoptosis in A549 cells. Chem Biol Interact. 181:154–167. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Zhao H, Liu J, Pan S, Sun Y, Li Q, Li F, Ma L and Guo Q: SOD mRNA and MDA expression in rectus femoris muscle of rats with different eccentric exercise programs and time points. PLoS One. 8:e736342013. View Article : Google Scholar : PubMed/NCBI

44 

Rakkola R, Matikainen S and Nyman TA: Proteome analysis of human macrophages reveals the upregulation of manganese-containing superoxide dismutase after toll-like receptor activation. Proteomics. 7:378–384. 2007. View Article : Google Scholar : PubMed/NCBI

45 

To EE, Broughton BR, Hendricks KS, Vlahos R and Selemidis S: Influenza A virus and TLR7 activation potentiate NOX2 oxidase-dependent ROS production in macrophages. Free Radic Res. 48:940–947. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Liu P, Jamaluddin M, Li K, Garofalo RP, Casola A and Brasier AR: Retinoic acid-inducible gene I mediates early antiviral response and Toll-like receptor 3 expression in respiratory syncytial virus-infected airway epithelial cells. J Virol. 81:1401–1411. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Huang S, Wei W and Yun Y: Upregulation of TLR7 and TLR3 gene expression in the lung of respiratory syncytial virus infected mice. Wei Sheng Wu Xue Bao. 49:239–245. 2009.PubMed/NCBI

48 

Groskreutz DJ, Monick MM, Powers LS, Yarovinsky TO, Look DC and Hunninghake GW: Respiratory syncytial virus induces TLR3 protein and protein kinase R, leading to increased double-stranded RNA responsiveness in airway epithelial cells. J Immunol. 176:1733–1740. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Rudd BD, Smit JJ, Flavell RA, Alexopoulou L, Schaller MA, Gruber A, Berlin AA and Lukacs NW: Deletion of TLR3 alters the pulmonary immune environment and mucus production during respiratory syncytial virus infection. J Immunol. 176:1937–1942. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Matsukura S, Kokubu F, Kurokawa M, Kawaguchi M, Ieki K, Kuga H, Odaka M, Suzuki S, Watanabe S, Takeuchi H, et al: Synthetic double-stranded RNA induces multiple genes related to inflammation through Toll-like receptor 3 depending on NF-kappaB and/or IRF-3 in airway epithelial cells. Clin Exp Allergy. 36:1049–1062. 2006. View Article : Google Scholar : PubMed/NCBI

51 

Ieki K, Matsukura S, Kokubu F, Kimura T, Kuga H, Kawaguchi M, Odaka M, Suzuki S, Watanabe S, Takeuchi H, et al: Double-stranded RNA activates RANTES gene transcription through co-operation of nuclear factor-kappaB and interferon regulatory factors in human airway epithelial cells. Clin Exp Allergy. 34:745–752. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Fink K, Duval A, Martel A, Soucy-Faulkner A and Grandvaux N: Dual role of NOX2 in respiratory syncytial virus- and sendai virus-induced activation of NF-kappaB in airway epithelial cells. J Immunol. 180:6911–6922. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Bsibsi M, Persoon-Deen C, Verwer RW, Meeuwsen S, Ravid R and Van Noort JM: Toll-like receptor 3 on adult human astrocytes triggers production of neuroprotective mediators. Glia. 53:688–695. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Larouche A, Berube P, Sarret P and Grignon S: Subacute H2O2, but not poly(IC), upregulates dopamine D2 receptors in retinoic acid differentiated SH-SY5Y neuroblastoma. Synapse. 62:70–73. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Grandvaux N, Soucy-Faulkner A and Fink K: Innate host defense: Nox and Duox on phox's tail. Biochimie. 89:1113–1122. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Geiler J, Michaelis M, Naczk P, Leutz A, Langer K, Doerr HW and Cinatl J Jr: N-acetyl-L-cysteine (NAC) inhibits virus replication and expression of pro-inflammatory molecules in A549 cells infected with highly pathogenic H5N1 influenza A virus. Biochem Pharmacol. 79:413–420. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Dong QG, Sclabas GM, Fujioka S, Schmidt C, Peng B, Wu T, Tsao MS, Evans DB, Abbruzzese JL, McDonnell TJ and Chiao PJ: The function of multiple IkappaB: NF-kappaB complexes in the resistance of cancer cells to Taxol-induced apoptosis. Oncogene. 21:6510–6519. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Ji K, Xing C, Jiang F, Wang X, Guo H, Nan J, Qian L, Yang P, Lin J, Li M, et al: Benzo[a]pyrene induces oxidative stress and endothelial progenitor cell dysfunction via the activation of the NF-κB pathway. Int J Mol Med. 31:922–930. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Wang XA, Zhang R, She ZG, Zhang XF, Jiang DS, Wang T, Gao L, Deng W, Zhang SM, Zhu LH, et al: Interferon regulatory factor 3 constrains IKKβ/NF-κB signaling to alleviate hepatic steatosis and insulin resistance. Hepatology. 59:870–885. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Spann KM, Tran KC and Collins PL: Effects of nonstructural proteins NS1 and NS2 of human respiratory syncytial virus on interferon regulatory factor 3, NF-kappaB, and proinflammatory cytokines. J Virol. 79:5353–5362. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Ling Z, Tran KC and Teng MN: Human respiratory syncytial virus nonstructural protein NS2 antagonizes the activation of beta interferon transcription by interacting with RIG-I. J Virol. 83:3734–3742. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Ren J, Liu T, Pang L, Li K, Garofalo RP, Casola A and Bao X: A novel mechanism for the inhibition of interferon regulatory factor-3-dependent gene expression by human respiratory syncytial virus NS1 protein. J Gen Virol. 92:2153–2159. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Wright PF, Karron RA, Madhi SA, Treanor JJ, King JC, O'Shea A, Ikizler MR, Zhu Y, Collins PL, Cutland C, et al: The interferon antagonist NS2 protein of respiratory syncytial virus is an important virulence determinant for humans. J Infect Dis. 193:573–581. 2006. View Article : Google Scholar : PubMed/NCBI

64 

Hosakote YM, Brasier AR, Casola A, Garofalo RP and Kurosky A: Respiratory syncytial virus infection triggers epithelial HMGB1 release as a damage-associated molecular pattern promoting a monocytic inflammatory response. J Virol. 90:9618–9631. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Hosakote YM, Jantzi PD, Esham DL, Spratt H, Kurosky A, Casola A and Garofalo RP: Viral-mediated inhibition of antioxidant enzymes contributes to the pathogenesis of severe respiratory syncytial virus bronchiolitis. Am J Respir Crit Care Med. 183:1550–1560. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Dave KA, Norris EL, Bukreyev AA, Headlam MJ, Buchholz UJ, Singh T, Collins PL and Gorman JJ: A comprehensive proteomic view of responses of A549 type II alveolar epithelial cells to human respiratory syncytial virus infection. Mol Cell Proteomics. 13:3250–3269. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Mochizuki H, Todokoro M and Arakawa H: RS virus-induced inflammation and the intracellular glutathione redox state in cultured human airway epithelial cells. Inflammation. 32:252–264. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Xu X, Zheng J, Zheng K, Hou Y, Zhao F and Zhao D: Respiratory syncytial virus NS1 protein degrades STAT2 by inducing SOCS1 expression. Intervirology. 57:65–73. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Hall CB, Douglas RG Jr, Simons RL and Geiman JM: Interferon production in children with respiratory syncytial, influenza, and parainfluenza virus infections. J Pediatr. 93:28–32. 1978. View Article : Google Scholar : PubMed/NCBI

70 

Roberts NJ Jr, Hiscott J and Signs DJ: The limited role of the human interferon system response to respiratory syncytial virus challenge: Analysis and comparison to influenza virus challenge. Microb Pathog. 12:409–414. 1992. View Article : Google Scholar : PubMed/NCBI

71 

Garofalo R, Mei F, Espejo R, Ye G, Haeberle H, Baron S, Ogra PL and Reyes VE: Respiratory syncytial virus infection of human respiratory epithelial cells up-regulates class I MHC expression through the induction of IFN-beta and IL-1 alpha. J Immunol. 157:2506–2513. 1996.PubMed/NCBI

72 

Jamaluddin M, Wang S, Garofalo RP, Elliott T, Casola A, Baron S and Brasier AR: IFN-beta mediates coordinate expression of antigen-processing genes in RSV-infected pulmonary epithelial cells. Am J Physiol Lung Cell Mol Physiol. 280:L248–L257. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Cheung MB, Sampayo-Escobar V, Green R, Moore ML, Mohapatra S and Mohapatra SS: Respiratory syncytial virus-infected mesenchymal stem cells regulate immunity via interferon beta and indoleamine-2,3-dioxygenase. PLoS One. 11:e01637092016. View Article : Google Scholar : PubMed/NCBI

74 

Hornung V, Schlender J, Guenthner-Biller M, Rothenfusser S, Endres S, Conzelmann KK and Hartmann G: Replication-dependent potent IFN-alpha induction in human plasmacytoid dendritic cells by a single-stranded RNA virus. J Immunol. 173:5935–5943. 2004. View Article : Google Scholar : PubMed/NCBI

75 

Schlender J, Hornung V, Finke S, Günthner-Biller M, Marozin S, Brzózka K, Moghim S, Endres S, Hartmann G and Conzelmann KK: Inhibition of toll-like receptor 7- and 9-mediated alpha/beta interferon production in human plasmacytoid dendritic cells by respiratory syncytial virus and measles virus. J Virol. 79:5507–5515. 2005. View Article : Google Scholar : PubMed/NCBI

76 

Guerrero-Plata A, Casola A, Suarez G, Yu X, Spetch L, Peeples ME and Garofalo RP: Differential response of dendritic cells to human metapneumovirus and respiratory syncytial virus. Am J Respir Cell Mol Biol. 34:320–329. 2006. View Article : Google Scholar : PubMed/NCBI

77 

Wang H, Peters N and Schwarze J: Plasmacytoid dendritic cells limit viral replication, pulmonary inflammation, and airway hyperresponsiveness in respiratory syncytial virus infection. J Immunol. 177:6263–6270. 2006. View Article : Google Scholar : PubMed/NCBI

78 

Al-Afif A, Alyazidi R, Oldford SA, Huang YY, King CA, Marr N, Haidl ID, Anderson R and Marshall JS: Respiratory syncytial virus infection of primary human mast cells induces the selective production of type I interferons, CXCL10, and CCL4. J Allergy Clin Immunol. 136:1346–1354.e1. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Kulka M, Alexopoulou L, Flavell RA and Metcalfe DD: Activation of mast cells by double-stranded RNA: Evidence for activation through Toll-like receptor 3. J Allergy Clin Immunol. 114:174–182. 2004. View Article : Google Scholar : PubMed/NCBI

80 

Hillyer P, Mane VP, Chen A, Dos Santos MB, Schramm LM, Shepard RE, Luongo C, Le Nouën C, Huang L, Yan L, et al: Respiratory syncytial virus infection induces a subset of types I and III interferons in human dendritic cells. Virology. 504:63–72. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 18 Issue 2

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
Wang MM, Lu M, Zhang CL, Wu X, Chen JX, Lv WW, Sun T, Qiu H and Huang SH: Oxidative stress modulates the expression of toll‑like receptor 3 during respiratory syncytial virus infection in human lung epithelial A549 cells. Mol Med Rep 18: 1867-1877, 2018
APA
Wang, M., Lu, M., Zhang, C., Wu, X., Chen, J., Lv, W. ... Huang, S. (2018). Oxidative stress modulates the expression of toll‑like receptor 3 during respiratory syncytial virus infection in human lung epithelial A549 cells. Molecular Medicine Reports, 18, 1867-1877. https://doi.org/10.3892/mmr.2018.9089
MLA
Wang, M., Lu, M., Zhang, C., Wu, X., Chen, J., Lv, W., Sun, T., Qiu, H., Huang, S."Oxidative stress modulates the expression of toll‑like receptor 3 during respiratory syncytial virus infection in human lung epithelial A549 cells". Molecular Medicine Reports 18.2 (2018): 1867-1877.
Chicago
Wang, M., Lu, M., Zhang, C., Wu, X., Chen, J., Lv, W., Sun, T., Qiu, H., Huang, S."Oxidative stress modulates the expression of toll‑like receptor 3 during respiratory syncytial virus infection in human lung epithelial A549 cells". Molecular Medicine Reports 18, no. 2 (2018): 1867-1877. https://doi.org/10.3892/mmr.2018.9089