Open Access

MicroRNA‑17 contributes to the suppression of the inflammatory response in lipopolysaccharide‑induced acute lung injury in mice via targeting the toll‑like receptor 4/nuclear factor‑κB pathway

  • Authors:
    • Shan Fu
  • View Affiliations

  • Published online on: May 12, 2020     https://doi.org/10.3892/ijmm.2020.4599
  • Pages: 131-140
  • Copyright: © Fu . 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

Acute lung injury (ALI) is a common lung disease with a high mortality rate, which is characterized by an excessive uncontrolled inflammatory response. MicroRNA (miR)‑17 has previously emerged as a novel regulatory molecule of inflammatory response in various complex diseases; however, the anti‑inflammatory action and associated molecular mechanisms of miR‑17 in ALI have not been fully elucidated. The aim of the present study was to investigate the role of miR‑17 in the inflammatory response in ALI and to elucidate the potential underlying mechanism. Using a lipopolysaccharide (LPS)‑induced ALI mouse model, it was observed that miR‑17 was significantly downregulated in lung tissues compared with the control group. In this model, ectopic expression of miR‑17 attenuated lung pathological damage, reduced lung wet/dry ratio and lung permeability, and increased survival rate in ALI mice. In addition, agomiR‑17 injection significantly suppressed LPS‑induced inflammation, as evidenced by a reduction in the activity of myeloperoxidase and the production of interleukin (IL)‑6, IL‑1β and tumor necrosis factor‑α in lung tissues. Of note, toll‑like receptor (TLR) 4, an upstream regulator of the nuclear factor (NF)‑κB inflammatory signaling pathway, was directly targeted by miR‑17, and its translation was suppressed by miR‑17 in vitro and in vivo. Using an LPS‑induced RAW264.1 macrophage injury model, it was observed that miR‑17 overexpression suppressed the pro‑inflammatory effect of LPS, while these inhibitory effects were markedly abrogated by TLR4 overexpression. In addition, TLR4 knockdown by si‑TLR4 mimicked the effects of miR‑17 overexpression on LPS‑induced cytokine secretion in the in vitro model. Further experiments revealed that miR‑17 significantly reduced the expression of key proteins in the NF‑κB pathway, including IKKβ, p‑IκBα and nuclear p‑p65, and suppressed the NF‑κB activity in ALI mice. Collectively, these results indicated that miR‑17 protected mice against LPS‑induced lung injury via inhibiting inflammation by targeting the TLR4/NF‑κB pathway; therefore, miR‑17 may serve as a potential therapeutic target for ALI.

Introduction

Acute lung injury (ALI) is a severe clinical condition with a high mortality rate among critically ill patients (1,2). Although there have been significant advancements in the therapeutic strategies of ALI, the morbidity and mortality rates remains high (3,4). In recent years, it has become widely accepted that inflammation is an important pathological characteristic of ALI, and contributes to the initiation and development of ALI (5). Therefore, inhibition of the inflammatory response may be key to the improvement of ALI patient outcomes.

It is well known that toll-like receptors (TLRs) play an important role in the inflammatory process triggered by lipopolysaccharide (LPS) (6,7). After TLR4 recognizes LPS, a series of cascades, including myeloid differentiation factor 88 (MyD88), are initiated, followed by activation of the nuclear factor (NF)-κB pathway and the secretion of pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6, ultimately resulting in ALI (8). Once the TLR4 pathway is inhibited, the inflammatory response can be alleviated, thereby attenuating ALI. Extensive studies have demonstrated, using an LPS induced in vivo ALI model, that inhibition of the TLR4 pathway is beneficial in ALI (9,10). For example, Zhang et al reported that inhibition of the TLR4/NF-κB signaling pathway improved the oxidative stress and inflammatory response in the lung tissues of ALI rats (11). Therefore, suppression of the activation of the TLR4/NF-κB pathway may alleviate inflammation-induced ALI.

MicroRNAs (miRNAs) are a family of short non-coding RNAs (with a mean size of 22 nucleotides), which suppress target gene expression through either translation repression or RNA degradation (12). Accumulating evidence has demonstrated that miRNAs potentially contribute to the development of ALI via regulation of target genes (13-15). For example, Yang et al observed that miR-140-5p inhibited LPS-induced inflammatory response in ALI via blocking the TLR4 pathway (16). Ling et al demonstrated that miR-494 inhibition improved lung injury through suppressing the inflammatory response in ALI rats (17). miR-17, a member of the miR-17-92 cluster, has been found to play an important role in ameliorating inflammatory response, particularly pulmonary inflammation (18,19). More importantly, a recent study has identified decreased expression of miR-17 in ALI mice, and miR-17 negatively regulates lung FOXA1 expression, which plays an important role in ALI by promoting the apoptosis of alveolar type II epithelial cells in vitro and in vivo (20). However, the function of miR-17 in inflammatory response in ALI has yet to be fully elucidated.

In the present study, an in vivo mice model of ALI and an in vitro LPS-induced RAW264.7 cell injury model were established to investigate the role and underlying mechanism of action of miR-17 in the regulation of inflammation in ALI. The aim was to determine whether miR-17 may hold promise as a novel treatment target for the prevention and treatment of ALI.

Materials and methods

Ethics statement

The protocol of the present study was approved by the Ethics Committee of the Affiliated Hospital of Inner Mongolia University for Nationalities (permit no. 2018-0139). The mice were treated humanely, and all measures were undertaken to minimize animal suffering. The mice were monitored every 12 h over a period of 1 week for health and behavior. A humane endpoint was used in our experiments according to previous report (21). The specific signs used to determine the endpoint included: i) Loss of >25% body weight compared with the starting weight; ii) decreased food or water intake; iii) decreased mobility/activity, lethargy, rough hair coat. Sacrifice was performed by intraperitoneal injection of sodium pentobarbital (50 mg/kg) followed by cervical dislocation, and death was confirmed when no spontaneous breathing for 2-3 min and no blinking reflex were observed (22). No animals died before meeting these endpoints. All mice (n=60) were euthanized as mentioned above.

Animals

A total of 60 male BALB/c mice (6-8 weeks old, weighing 18-22 g) were obtained from the Shanghai SLAC Laboratory Animal Co. Ltd. BALB/c mice were housed under standard conditions (12-h light-dark cycle, 25-27°C, ~40% humidity) with free access to food and water throughout the duration of the experiments. A total of 20 mice were randomly divided into four groups (n=5/group) as follows: i) Control, ii) LPS, iii) LPS + agomir-17 and iv) LPS + agomir-negative control (NC) groups. LPS group mice were injected through the tail vein with 2 mg/kg LPS. The control group received the same volume of normal saline. Mice in the agomir-17 or agomir NC groups were injected intravenously with agomir-17 or agomir NC (8 mg/kg), respectively (20), 24 h prior to LPS induction. All mice were anesthetized with pentobarbital sodium (50 mg/kg, intraperitoneal injection), and euthanized by cervical dislocation at 24 h after LPS treatment (except those included in the survival experiment), then the lung tissues and bronchoalveolar lavage fluid (BALF) were collected for subsequent analysis.

For the survival rate analysis, 40 mice were divided into four groups (n=10/group) as mentioned above. The survival rate from 0 to 7 days was observed and calculated using the Kaplan-Meier method. Agomir-17/NC were designed and synthesized by RiboBio. The sequences were as follows: Agomir-17: 5′-CAA AGU GCU UAC AGU GCA GGU AG-3′; and agomir NC: 5′-GUC CUG AGA AGG CUA GCA UAG AU-3′.

Reverse transcription-quantitative PCR (RT-qPCR) analysis

Total RNA was isolated from lung tissues or cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). RT of miR-17 was performed using the miScript II RT kit (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 30 min. miR-17 expression was measured using the Exiqon SYBR Green Master Mix (Exiqon; Qiagen, Inc.) on a Light Cycler instrument (Bio-Rad Laboratories, Inc.). The thermocycling conditions were as follows: A hot start step at 95°C for 10 min, followed by 40 cycles at 95°C for 15 sec and 60°C for 1 min. The primers for RT-qPCR analysis were as follows: miR-17 forward, 5′-AGGCCCAAAGTGCTGTTCGT-3′ and reverse, 5′-GTGCAGGGTCCGAGGT-3′; U6 forward, 5′-TGC GGG TGC TCG CTT CGC AGC-3′ and reverse, 5′-CCA GTG CAG GGT CCG AGG T-3′. The miRNA relative expressions were analyzed using the 2−ΔΔCq method (23).

Lung histology

After a 24-h LPS challenge, the mice were sacrificed and the lung tissues were harvested and fixed in 10% formalin for 24 h at 4°C, embedded in paraffin, and cut into 4-µm sections. The sections were stained using hematoxylin for 5 min and rinsed with distilled water, followed by color separation with alcohol hydrochloric acid at room temperature. Subsequently, the samples were stained with eosin for 2 min, dehydrated, cleared, dried and mounted at room temperature. Histological changes were observed and photographed under a light microscope (E-800M, Nikon Corporation) at a magnification of ×200 and 400.

Evaluation of lung permeability

The Evans Blue (EB) dye extravasation method was used to assess pulmonary permeability as previously described (24). Briefly, EB dye (20 mg/kg, Sigma-Aldrich; Merck KGaA) at a concentration of 0.5% (5 mg/ml) in normal saline was injected into the mice of each group through the tail vein. After 2 h, the mice were sacrificed and then the dye was extracted by incubation in formamide for 24 h at 60°C. The light absorbance at 620 nm was measured, and the dye concentration in lung homogenate was calculated against a standard curve and was expressed as µg of Evans blue dye per g of lung tissue.

Lung wet/dry (W/D) ratio

The W/D ratio was used to assess pulmonary edema. After a 24-h LPS challenge, the right lung of the mice was harvested and immediately weighted, then dried in an incubator at 80°C for 60 h.

Measurement of IL-6, IL-1β and TNF-α

The supernatant from the BALF was collected by centrifugation at 12,000 x g for 10 min at 4°C. For cultured cells, the supernatant was carefully collected by centrifugation 12,000 × g for 10 min at 4°C. The concentrations of IL-6, IL-1β and TNF-α were analyzed by using IL-6 (cat. no. p1330), IL-1β (cat. no. p1305) and TNF-α (cat. no. pt518) ELISA kits, respectively, according to the instructions of the manufacturer (Beyotime Institute of Biotechnology).

Measurement of myeloperoxidase (MPO) activity

Lung tissue homogenate was subjected to MPO assay using a commercial kit (cat. no. ab105136, Abcam) and the absorbance at 460 nm was detected using a microplate reader (Bio-Tek Instruments, Inc.).

Cell culture and transfection

RAW264.7 cells were obtained from Sciencell Research Laboratories and maintained in DMEM supplemented with 10% FBS (Sigma-Aldrich; Merck KGaA), 1% penicillin and streptomycin (Sigma-Aldrich; Merck KGaA) at 37°C and 5% CO2 in an incubator.

After RAW264.7 cells in 6-well plates had grown to ~80% confluence, 20 nM miR-17 mimics, 20 nM miR-17 inhibitor, 2 µg pcDNA-TLR4 or 30 nM si-TLR4 were transfected into the cells at 37°C for 24 h using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). miR-17 inhibitor, miR-17 mimics and the corresponding control vectors were purchased from RiboBio Co., Ltd. The TLR4 overexpressing vector pcDNA-TLR4 and empty vector pcDNA were constructed by Qiagen, Inc. In addition, TLR4 siRNA (si-TLR4) and corresponding negative control siRNA (si-Scramble) were purchased from RiboBio Co., Ltd. The sequences were as follows: miR-17-5p mimic: 5′-CAA AGU GCU UAC AGU GCA GGU AG-3′; mimics NC: 5′-GUC CUG AGA AGG CUA GCA UAG AU-3′; miR-17-5p inhibitor: 5′-CUA CCU GCA CUG UAA GCA CUU UG-3′, inhibitor NC 5′-CUA UGC U AGC CUU CUC AGG ACU U-3′.

Cell proliferation

The effect of LPS (2 µg/ml) on RAW264.7 cells was measured by using the MTT assay. At the end of the transfection, 20 µl MTT solution (Sigma-Aldrich; Merck KGaA) was added into each well (2x105/well), and RAW264.7 cells were cultured for another 2 h. Then, the absorbance at 450 nm was detected by a microplate reader (Bio-Tek Instruments, Inc.).

Luciferase assay

miRNA target prediction tools, including PicTar version 2007 (https://pictar.mdc-berlin.de/) and TargetScan Release 7.0 (http://targetscan.org/) were used to search for the putative targets of miR-17. The dual luciferase reporter assay was performed as described previously (25). RAW264.7 cells were treated with miR-17 mimics, miR-17 inhibitor the luciferase reporter plasmids (wt-TLR4-UTR-pGL3 or mt-TLR4-UTR-pGL3), and pRL-TK-Renilla control plasmid (Promega Corporation) using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). At 48 h post-transfection, lucif-erase activity was detected using the Dual Luciferase Reporter kit (Beyotime Institute of Biotechnology).

Western blot analysis

Protein was extracted from the mouse lung tissues using the Nuclear and Cytoplasmic Protein Extraction kit (Beyotime Institute of Biotechnology). The protein concentration was measured using a protein analysis kit (Bio-Rad Laboratories, Inc.). Extracted protein samples (40 µg) were separated by 12% SDS-PAGE (w/v) and transferred onto a PVDF membrane (EMD Millipore). The membrane was blocked with 5% skimmed milk for 2 h at room temperature, followed by incubation with primary antibodies against TLR4 (cat. no. 14358, 1:2,000 dilution), nuclear p-p65 (Ser-468, cat. no. 3039, 1:1,000 dilution), p-IκB-α (Ser-32, cat. no. 2859, 1:1,000 dilution), IκB-α (cat. no. 4814, 1:1,000 dilution), Histone H3 (cat. no. 9728, 1:1,000 dilution) and β-actin (cat. no. 3700, 1:1,000) (all from Cell Signaling Technology, Inc.) at 4°C overnight, followed by HRP-conjugated goat anti-rabbit IgG (1:10,000; cat. no. 205718; Abcam). β-actin and Histone H3 served as internal controls. The protein bands were developed using an ECL kit (GE Healthcare) and blot bands were quantified with ImageJ version 1.46 (Rawak Software, Inc.).

Statistical analysis

GraphPad Prism 5.0 (GraphPad Software, Inc.) was used to perform the statistical analyses. When only two groups were compared, Student's t-test was used. One-way analysis of variance followed by Tukey's post hoc test was applied to compare differences between multiple groups. Survival analysis was performed using the Kaplan-Meier method. All data are presented as the mean ± standard deviation. P<0.05 was considered to indicate statistically significant differences.

Results

miR-17 is downregulated in lung tissues of ALI mice

First, an LPS-induced ALI mouse model was established, which is widely used to simulate the pathological conditions of severe lung injury in vivo (26). Following LPS treatment, the pathological changes of lung tissues, lung W/D ratio (an indicator of lung edema), pulmonary capillary permeability, MPO activity and the inflammatory response were evaluated. As shown in Fig. 1A, the H&E staining results revealed that LPS treatment caused obvious pathological changes, including inflammatory cell infiltration and widespread increased alveolar wall thickness, compared with the control group (n=5/group). The W/D ratio of mice in the LPS group (n=5/group) was markedly higher compared with that in the control group (Fig. 1B). The pulmonary capillary permeability was measured by the EB dye method (n=5/group) and the results demonstrated that LPS treatment led to a significant increase in EB dye extravasation compared with the control group (Fig. 1C). The MPO activity, determined by a commercial assay (n=5/group), was markedly upregulated following LPS stimulation compared with that in the control group (Fig. 1D). Furthermore, the expression of IL-1β, IL-6 and TNF-α was measured by ELISA (n=5/group). As shown in Fig. 1E, the levels of these cytokines were obviously increased in the LPS group compared with those in the control group (Fig. 1E). All these data indicated that the LPS-induced ALI mouse model was successfully constructed.

To investigate the potential involvement of miR-17 in ALI, the expression level of miR-17 was measured in lung tissues from ALI mice (n=10/group). It was observed that miR-17 was markedly downregulated in the lung tissues from ALI mice (Fig. 1F), suggesting that miR-17 may play an important role in the pathogenesis of ALI.

Overexpression of miR-17 improves LPS-induced ALI in vivo

To further examine the therapeutic effect of miR-17 in LPS-induced ALI, agomiR-17 was intravenously administered to the mice, followed by LPS treatment. The miRNA transfection efficiency was first evaluated using RT-qPCR. The results demonstrated that the miR-17 expression in lung tissues from ALI mice was increased following agomiR-17 injection (n=5/group; Fig. 2A). Subsequently, the pathological changes, pulmonary edema, pulmonary capillary permeability and MPO activity were determined with the use of H&E staining, EB extravasation, W/D ratio and MPO assays, respectively (n=5/group). It was observed that LPS treatment was associated with a higher extent of lung injury compared with the control group, while agomir-17 alleviated the severity and distribution of lung lesions caused by LPS (Fig. 2B). As shown in Fig. 2C and D, agomir-17 treatment markedly reduced W/D ratio and EB extravasation in ALI mice, suggesting that miR-17 effectively improved the pulmonary edema and pulmonary capillary permeability. It was also observed that the increased MPO activity induced by LPS was obviously inhibited by agomir-17 (Fig. 2E). In addition, the Kaplan-Meier method revealed that 50% of mice in the LPS group died within 7 days, whereas the 7-day survival rate after agomiR-17 injection was markedly higher compared with the LPS group (n=10/group; Fig. 2F). More importantly, the protein levels of cytokines, including IL-1β, IL-6 and TNF-α, induced by LPS were significantly decreased following agomir-17 injection (n=5/group; Fig. 2G). Collectively, these data indicate that miR-17 upregulation may improve LPS-induced ALI.

TLR4 is a direct target of miR-17

To investigate the potential molecular mechanism of miR-17-regulated inflammatory events in ALI, TargetScan and PicTar analyses were conducted to predict the target genes of miR-17. As shown in Fig. 3A and B, TLR4 was identified as a potential target gene of miR-17, with the target site located in the 3′-UTR of TLR4 mRNA. It is well known that TLR-4 is a common receptor of LPS, and its downstream signaling effector, NF-κB, is closely associated with inflammatory response in ALI (14,27-30). Thus, TLR4 was selected for the subsequent investigation. The potential targeting interaction between miR-17 and TLR-4 was verified by dual luciferase reporter assays. The results demonstrated that the miR-17 mimics significantly inhibited the luciferase activity of the TLR4-3′UTR wt, and that miR-17 inhibitor transfection resulted in a significant increase in luciferase activity; however, no changes were observed in the cells following co-transfection of TLR4 3′-UTR-mut with either miR-17 mimics or inhibitor (Fig. 3C). miR-17 was then introduced into RAW264.1 cells and the expression of TLR4 was analyzed by western blotting. It was observed that miR-17 overexpression suppressed the protein expression of TLR4, while miR-17 inhibition promoted TLR4 expression in RAW264.1 macrophages (Fig. 3D). Of note, the protein expression of TLR4 was also found to be decreased in lung tissues of ALI mice after agomiR-17 injection (n=5/group; Fig. 3E). These results indicate that TLR4 may be a functional target of miR-17 against lung inflammatory response in ALI mice.

miR-17 is downregulated in LPS-treated RAW264.1 macrophages

In the physiopathology of ALI, macrophages play an important role in the regulation of inflammation (31,32). To further confirm whether miR-17 is involved in LPS-mediated immune response, RAW264.1 macrophages were treated with LPS at different concentrations (0-2.5 µg/ml) for different times (0-36 h), as previously described (33). After LPS treatment of RAW264.1 cells, the expression level of miR-17 decreased in a dose-dependent manner (Fig. 4A). Furthermore, it was also observed that miR-17 expression time-dependently decreased, while the expression of miR-17 reached a nadir at 12 h after LPS treatment (Fig. 4B). Additionally, the MTT assay was used to assess the effect of LPS (2 µg/ml) on the viability of RAW264.7 cells, and the results demonstrated that cell viability was not affected by LPS at any of the different time points (Fig. 4C). Based on the data mentioned above, treatment with 2 µg/ml LPS for 12 h was selected as the optimal conditions for subsequent experiments.

Overexpression of miR-17 improves the inflammatory response by targeting TLR4 in LPS-treated RAW264.7 cells

As mentioned above, TLR4 was found to be a direct target of miR-17 in RAW264.7 cells; therefore, it was further investigated whether miR-17 regulates the inflammatory response in ALI through targeting TLR4 by co-transfecting miR-17 mimics and pcDNA-TLR4 into RAW264.7 cells, followed by treatment with LPS. Consistently with the results of the inflammatory cytokine generation in vivo, LPS treatment resulted in significant increases in the levels of IL-1β, IL-6 and TNF-α, whereas these effects caused by LPS were attenuated by overexpression of miR-17. Interestingly, TLR4 overexpression reversed the inhibitory effects of miR-17 overexpression on the expression of these cytokines in this cell model (Fig. 5A-C). In addition, RAW264.7 cells were transfected with si-TLR4 to determine the role of TLR4 in the regulation of inflammatory response. As shown in Fig. 5D-F, the production of these cytokines induced by LPS was markedly reduced by TLR4 knockdown, which was similar to the effect of miR-17 mimics on LPS-treated RAW264.7 cells. These findings demonstrated that miR-17 improves the inflammatory response by targeting TLR4 in an ALI cell model.

Overexpression of miR-17 blocks the activation of the NF-κB signaling pathway

According to previous research, TLR4 is a common receptor of LPS, and its downstream signaling effector, the NF-κB pathway, plays a crucial role in the inflammatory response in ALI (34,35). Therefore, to further investigate the possible mechanisms underlying the role of miR-17 in the inflammatory response, we focused on the phosphorylation/activation of the NF-κB signaling pathway in ALI mice using western blot assay (n=5/group). The data demonstrated that LPS treatment significantly upregulated the levels of p-IκBα and nuclear p-p65 compared with the control group. However, compared with the LPS group, the levels of these proteins were markedly downregulated in the LPS + agomiR-17 group (Fig. 6A). In addition, it was also observed that LPS led to a significant increase in NF-κB activity, whereas agomiR-17 markedly reduced the increased NF-κB activity caused by LPS (Fig. 6B). These results demonstrated that miR-17 exerts its potent inhibitory effects against LPS-induced lung injury via the NF-κB signaling pathway.

Discussion

In the present study, miR-17 was significantly downregulated in both LPS-induced ALI mice and LPS-treated RAW264.7 cells. Further analysis revealed that miR-17 improved LPS-induced lung injury through inhibition of the inflammatory response through blocking the activation of the TLR4/NF-κB pathway. These results suggest that miR-17 may be of value as a potential therapeutic approach to ALI.

Accumulating evidence has demonstrated that miRNAs participate in the regulation of the inflammatory response in several types of diseases (36,37). A number of miRNAs, such as miR-9, miR-147 and miR-132, have been reported to exert an inhibitory effect on inflammatory response (38-40). Rao et al demonstrated that miR-17 attenuated staphylococcal enterotoxin B-induced lung injury via suppressing the pulmonary inflammatory response (41). Wang and Zhang found that miR-17 overexpression mediated the protective effects of resveratrol against LPS-induced cell injury through reducing the production of pro-inflammatory cytokines in the human keratinocyte cell line HaCaT (18). However, the mechanism underlying the association between miR-17 and LPS-induced ALI remains elusive. Therefore, it is necessary to further investigate whether miR-17 affects the inflammatory response in ALI. The present study demonstrated that the expression of miR-17 was low in the lung tissues of ALI mice, whereas agomiR-17 injection reduced tissue damage and lung edema, and inhibited the LPS-induced inflammatory response in an LPS-induced ALI mouse model. Furthermore, the results of the present study demonstrated that agomiR-17 markedly improved the survival rate of mice with LPS-induced ALI. Using an in vitro RAW264.7 macrophage model, downregulation of miR-17 in RAW264.7 cells was observed after LPS stimulation. Consistently with the results in vivo, this demonstrated that miR-17 can attenuate LPS-induced inflammatory response. Collectively, these data suggest that upregulation of miR-17 may improve LPS-induced ALI by inhibition of the inflammatory response.

TLR4 is an essential LPS signaling receptor, which is known to play an important role in inflammatory response in animal or cell models of ALI (42,43). For example, Hu et al reported that the inhibition of the TLR4 signaling pathway suppressed the production of inflammatory cytokines, thereby improving ALI in rats (28). He et al demonstrated that the LPS/TLR4 axis can activate the NF-κB signaling pathway, resulting in inflammatory cell infiltration (44). A previous study also reported that miR-27a alleviated LPS-induced ALI in mice via inhibiting inflammation through modulating the TLR4/MyD88/NF-κB pathway (45). Interestingly, the expression levels of TLR4 were previously reported to be regulated by miR-17 in an LPS-induced sepsis mouse model (46); however, it is ambiguous whether miR-17 serves as a protective factor in LPS-induced ALI through regulation of TLR4. In the present study, TLR4 was identified as a target of miR-17. Furthermore, TLR4 overexpression abrogated the inhibitory effects of miR-17 on inflammatory response in LPS-induced RAW 264.7 cells. By contrast, silencing TLR4 by siTLR4 had a similar effect to that of miR-17 on LPS-treated RAW 264.7 cells. Collectively, these findings indicate that the miR-17/TLR4 axis may serve as a novel therapeutic target for LPS-induced ALI.

Activation of canonical NF-κB plays important roles in LPS-induced ALI (47,48). In ALI, NF-κB is constitutively activated and is involved in promoting the inflammatory response, which suggests that inhibiting the activity of NF-κB may constitute a promising therapeutic approach to ALI (49). According to previous research, TLR4 is a recognized inducer of the NF-κB signaling pathway, and it has been demonstrated that inhibition of TLR-4/NF-κB signaling may improve LPS-induced ALI in a mouse model (50,51). Therefore, the mechanism through which miR-17 affects the TLR-4/NF-κB signaling pathway requires further investigation. In the present study, it was observed that the overexpression of miR-17 significantly suppressed the LPS-induced activation of NF-κB in vivo. These results indicate that miR-17 may alleviate the LPS-induced inflammatory response by inhibiting the TLR4/NF-κB signaling pathway.

In conclusion, the present study demonstrated that miR-17 can alleviate LPS-induced ALI in mice through inhibiting the inflammatory response via the TLR4/NF-κB signaling pathway. Therefore, the miR-17/TLR4 axis may be a candidate target for the treatment of patients with ALI.

Funding

No funding was received.

Availability of data and materials

All data generated and/or analyzed during the present study are included in this published article.

Authors' contributions

SF conceptualized the study design, obtained the experimental materials, performed the experiments, analyzed the data and wrote the paper. The author has read and approved the final version of the manuscript.

Ethics approval and consent to participate

All individuals provided informed consent for the use of human specimens for clinical research. The present study was approved by the Affiliated Hospital of Inner Mongolia University for the Nationalities Ethics Committees.

Patient consent for publication

Not applicable.

Competing interests

The author declares that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Herridge MS, Tansey CM, Matté A, Tomlinson G, Diaz-Granados N, Cooper A, Guest CB, Mazer CD, Mehta S, Stewart TE, et al: Functional disability 5 years after acute respiratory distress syndrome. N Engl J Med. 364:1293–1304. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Mendez JL and Hubmayr RD: New insights into the pathology of acute respiratory failure. Curr Opin Crit Care. 11:29–36. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Zhang J, Cao J, Feng J, Wu Q and Chen BY: A study of noninvasive positive-pressure mechanical ventilation in the treatment of acute lung injury with a complex critical care ventilator. J Int Med Res. 42:788–798. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Zhang Z, Chen L and Ni H: The effectiveness of Corticosteroids on mortality in patients with acute respiratory distress syndrome or acute lung injury: A secondary analysis. Sci Rep. 5:176542015. View Article : Google Scholar : PubMed/NCBI

5 

Deng X, Jin K, Li Y, Gu W, Liu M and Zhou L: Platelet-derived growth factor and transforming growth factor β1 regulate ARDS-associated lung fibrosis through distinct signaling pathways. Cell Physiol Biochem. 36:937–946. 2015. View Article : Google Scholar

6 

Wang X, Zhang L, Duan W, Liu B, Gong P, Ding Y and Wu X: Anti-inflammatory effects of triptolide by inhibiting the NF-κB signalling pathway in LPS-induced acute lung injury in a murine model. Mol Med Rep. 10:447–452. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, Takeda K and Akira S: Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: Evidence for TLR4 as the Lps gene product. J Immunol. 162:3749–3752. 1999.PubMed/NCBI

8 

Chang X, He H, Zhu L, Gao J, Wei T, Ma Z and Yan T: Protective effect of apigenin on Freund's complete adjuvant-induced arthritis in rats via inhibiting P2X7/NF-κB pathway. Chem Biol Interact. 236:41–46. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Wei D and Huang Z: Anti-inflammatory effects of triptolide in LPS-induced acute lung injury in mice. Inflammation. 37:1307–1316. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Jiang Q, Yi M, Guo Q, Wang C, Wang H, Meng S, Liu C, Fu Y, Ji H and Chen T: Protective effects of polydatin on lipopolysac-charide-induced acute lung injury through TLR4-MyD88-NF-κB pathway. Int Immunopharmacol. 29:370–376. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Zhang ZM, Wang YC, Chen L and Li Z: Protective effects of the suppressed NF-κB/TLR4 signaling pathway on oxidative stress of lung tissue in rat with acute lung injury. Kaohsiung J Med Sci. 35:265–276. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Tao Z, Yuan Y and Liao Q: Alleviation of lipopolysac-charides-induced acute Lung injury by MiR-454. Cell Physiol Biochem. 38:65–74. 2016. View Article : Google Scholar

14 

Yang Y, Yang F, Yu X, Wang B, Yang Y and Zhou X, Cheng R, Xia S and Zhou X: miR-16 inhibits NLRP3 inflammasome activation by directly targeting TLR4 in acute lung injury. Biomed Pharmacother. 112:1086642019. View Article : Google Scholar : PubMed/NCBI

15 

Zhou T, Garcia JG and Zhang W: Integrating microRNAs into a system biology approach to acute lung injury. Transl Res. 157:180–190. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Yang Y, Liu D, Xi Y and Li J, Liu B and Li J: Upregulation of miRNA-140-5p inhibits inflammatory cytokines in acute lung injury through the MyD88/NF-κB signaling pathway by targeting TLR4. Exp Ther Med. 16:3913–3920. 2018.PubMed/NCBI

17 

Ling Y, Li ZZ, Zhang JF, Zheng XW, Lei ZQ, Chen RY and Feng JH: MicroRNA-494 inhibition alleviates acute lung injury through Nrf2 signaling pathway via NQO1 in sepsis-associated acute respiratory distress syndrome. Life Sci. 210:1–8. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Wang X and Zhang Y: Resveratrol alleviates LPS-induced injury in human keratinocyte cell line HaCaT by up-regulation of miR-17. Biochem Biophys Res Commun. 501:106–112. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Oglesby IK, Vencken SF, Agrawal R, Gaughan K, Molloy K, Higgins G, McNally P, McElvaney NG, Mall MA and Greene CM: miR-17 overexpression in cystic fibrosis airway epithelial cells decreases interleukin-8 production. Eur Respir J. 46:1350–1360. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Xu Z, Zhang C, Cheng L, Hu M, Tao H and Song L: The microRNA miR-17 regulates lung FoxA1 expression during lipopolysaccharide-induced acute lung injury. Biochem Biophys Res Commun. 445:48–53. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Toyama M, Kudo D, Aoyagi T, Miyasaka T, Ishii K, Kanno E, Kaku M, Kushimoto S and Kawakami K: Attenuated accumulation of regulatory T cells and reduced production of interleukin 10 lead to the exacerbation of tissue injury in a mouse model of acute respiratory distress syndrome. Microbiol Immunol. 62:111–123. 2018. View Article : Google Scholar

22 

Wen N, Guo B, Zheng H, Xu L, Liang H, Wang Q, Wang D, Chen X, Zhang S, Li Y and Zhang L: Bromodomain inhibitor jq1 induces cell cycle arrest and apoptosis of glioma stem cells through the VEGF/PI3K/AKT signaling pathway. Int J Oncol. 55:879–895. 2019.PubMed/NCBI

23 

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

24 

Duan Y, Learoyd J, Meliton AY, Leff AR and Zhu X: Inhibition of Pyk2 blocks lung inflammation and injury in a mouse model of acute lung injury. Respir Res. 13:42012. View Article : Google Scholar : PubMed/NCBI

25 

Yao Y, Sun F and Lei M: miR-25 inhibits sepsis-induced cardiomyocyte apoptosis by targetting PTEN. Biosci Rep. 38:BSR201715112018. View Article : Google Scholar : PubMed/NCBI

26 

Cai ZG, Zhang SM, Zhang Y, Zhou YY, Wu HB and Xu XP: MicroRNAs are dynamically regulated and play an important role in LPS-induced lung injury. Can J Physiol Pharmacol. 90:37–43. 2012. View Article : Google Scholar

27 

Dong Z and Yuan Y: Accelerated inflammation and oxidative stress induced by LPS in acute lung injury: Iotanhibition by ST1926. Int J Mol Med. 41:3405–3421. 2018.PubMed/NCBI

28 

Hu X, Liu S, Zhu J and Ni H: Dachengqi decoction alleviates acute lung injury and inhibits inflammatory cytokines production through TLR4/NF-κB signaling pathway in vivo and in vitro. J Cell Biochem. 120:8956–8964. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Liu JX, Li X, Yan FG, Pan QJ, Yang C, Wu MY, Li G and Liu HF: Protective effect of forsythoside B against lipo-polysaccharide-induced acute lung injury by attenuating the TLR4/NF-κB pathway. Int Immunopharmacol. 66:336–346. 2019. View Article : Google Scholar

30 

Deng G, He H, Chen Z, OuYang L, Xiao X, Ge J, Xiang B, Jiang S and Cheng S: Lianqinjiedu decoction attenuates LPS-induced inflammation and acute lung injury in rats via TLR4/NF-κB pathway. Biomed Pharmacother. 96:148–152. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Zeng Z, Gong H, Li Y, Jie K, Ding C, Shao Q, Liu F, Zhan Y, Nie C, Zhu W and Qian K: Upregulation of miR-146a contributes to the suppression of inflammatory responses in LPS-induced acute lung injury. Exp Lung Res. 39:275–282. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Zhu WD, Xu J, Zhang M, Zhu TM, Zhang YH and Sun K: MicroRNA-21 inhibits lipopolysaccharide-induced acute lung injury by targeting nuclear factor-κB. Exp Ther Med. 16:4616–4622. 2018.PubMed/NCBI

33 

Jiang K, Guo S, Zhang T, Yang Y, Zhao G, Shaukat A, Wu H and Deng G: Downregulation of TLR4 by miR-181a provides negative feedback regulation to lipopolysaccharide-induced inflammation. Front Pharmacol. 9:1422018. View Article : Google Scholar : PubMed/NCBI

34 

Yu YL, Yu G, Ding ZY, Li SJ and Fang QZ: Overexpression of miR-145-5p alleviated LPS-induced acute lung injury. J Biol Regul Homeost Agents. 33:1063–1072. 2019.PubMed/NCBI

35 

Wang B, Wang J, Lu D, Qi N and Liu Q: The defensive action of LYRM03 on LPS-induced acute lung injury by NF-κB/TLR4/NLRP3 signals. J Invest Surg. 1–13. 2019.

36 

Li W, Qiu X, Jiang H, Han Y, Wei D and Liu J: Downregulation of miR-181a protects mice from LPS-induced acute lung injury by targeting Bcl-2. Biomed Pharmacother. 84:1375–1382. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Wei C, Li L, Kim IK, Sun P and Gupta S: NF-κB mediated miR-21 regulation in cardiomyocytes apoptosis under oxidative stress. Free Radic Res. 48:282–291. 2014. View Article : Google Scholar

38 

Zhou Z and You Z: Mesenchymal stem cells alleviate LPS-induced acute lung injury in mice by MiR-142a-5p-controlled pulmonary endothelial cell autophagy. Cell Physiol Biochem. 38:258–266. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Lee HM, Kim TS and Jo EK: MiR-146 and miR-125 in the regulation of innate immunity and inflammation. BMB Rep. 49:311–318. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Bao MH, Li JM, Luo HQ, Tang L, Lv QL, Li GY and Zhou HH: NF-κB-regulated miR-99a modulates endothelial cell inflammation. Mediators Inflamm. 2016:53081702016. View Article : Google Scholar

41 

Rao R, Nagarkatti PS and Nagarkatti M: Δ(9) Tetrahydrocannabinol attenuates Staphylococcal enterotoxin B-induced inflammatory lung injury and prevents mortality in mice by modulation of miR-17-92 cluster and induction of T-regulatory cells. Br J Pharmacol. 172:1792–1806. 2015. View Article : Google Scholar :

42 

Lu YC, Yeh WC and Ohashi PS: LPS/TLR4 signal transduction pathway. Cytokine. 42:145–151. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Meng L, Li L, Lu S, Li K, Su Z, Wang Y, Fan X, Li X and Zhao G: The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-κB and PI3K/Akt/mTOR pathways. Mol Immunol. 94:7–17. 2018. View Article : Google Scholar

44 

He X, Qian Y, Li Z, Fan EK, Li Y, Wu L, Billiar TR, Wilson MA, Shi X and Fan J: TLR4-upregulated IL-1β and IL-1RI promote alveolar macrophage pyroptosis and lung inflammation through an autocrine mechanism. Sci Rep. 6:316632016. View Article : Google Scholar

45 

Ju M, Liu B, He H, Gu Z, Liu Y, Su Y, Zhu D, Cang J and Luo Z: MicroRNA-27a alleviates LPS-induced acute lung injury in mice via inhibiting in fl ammation and apoptosis through modulating TLR4/MyD88/NF-κB pathway. Cell cycle. 17:2001–2018. 2018. View Article : Google Scholar :

46 

Ji ZR, Xue WL and Zhang L: Schisandrin B attenuates inflammation in LPS-induced sepsis through miR-17-5pdownregulating TLR4. Inflammation. 42:731–739. 2019. View Article : Google Scholar

47 

Bollenbach M, Salvat E, Daubeuf F, Wagner P, Yalcin I, Humo M, Letellier B, Becker LJ, Bihel F, Bourguignon JJ, et al: Phenylpyridine-2-ylguanidines and rigid mimetics as novel inhibitors of TNFα overproduction: Beneficial action in models of neuropathic pain and of acute lung inflammation. Eur J Med Chem. 147:163–182. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Lorne E, Dupont H and Abraham E: Toll-like receptors 2 and 4: Initiators of non-septic inflammation in critical care medicine? Intensive Care Med. 36:1826–1835. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Narukawa M: Physiological responses to taste signals of functional food components. Biosci Biotechnol Biochem. 82:200–206. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Wang J, Fan SM and Zhang J: Epigallocatechin-3-gallate ameliorates lipopolysaccharide-induced acute lung injury by suppression of TLR4/NF-kappaB signaling activation. Braz J Med Biol Res. 52:e80922019. View Article : Google Scholar

51 

Wang M, Niu J, Ou L, Deng B, Wang Y and Li S: Zerumbone protects against carbon tetrachloride (CCl4)-induced acute liver injury in mice via inhibiting oxidative stress and the inflammatory response: Involving the TLR4/NF-κB/COX-2 pathway. Molecules. 24:E19642019. View Article : Google Scholar

Related Articles

Journal Cover

July-2020
Volume 46 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fu S: MicroRNA‑17 contributes to the suppression of the inflammatory response in lipopolysaccharide‑induced acute lung injury in mice via targeting the toll‑like receptor 4/nuclear factor‑κB pathway. Int J Mol Med 46: 131-140, 2020
APA
Fu, S. (2020). MicroRNA‑17 contributes to the suppression of the inflammatory response in lipopolysaccharide‑induced acute lung injury in mice via targeting the toll‑like receptor 4/nuclear factor‑κB pathway. International Journal of Molecular Medicine, 46, 131-140. https://doi.org/10.3892/ijmm.2020.4599
MLA
Fu, S."MicroRNA‑17 contributes to the suppression of the inflammatory response in lipopolysaccharide‑induced acute lung injury in mice via targeting the toll‑like receptor 4/nuclear factor‑κB pathway". International Journal of Molecular Medicine 46.1 (2020): 131-140.
Chicago
Fu, S."MicroRNA‑17 contributes to the suppression of the inflammatory response in lipopolysaccharide‑induced acute lung injury in mice via targeting the toll‑like receptor 4/nuclear factor‑κB pathway". International Journal of Molecular Medicine 46, no. 1 (2020): 131-140. https://doi.org/10.3892/ijmm.2020.4599