MicroRNA‑421 attenuates macrophage‑mediated inflammation by inhibiting PDCD4 in vitro

  • Authors:
    • Huifang Liu
    • Junzhi Sun
    • Li Gao
    • Lingling Fan
    • Dongmei Chen
    • Lifang Wu
  • View Affiliations

  • Published online on: May 25, 2021     https://doi.org/10.3892/mmr.2021.12166
  • Article Number: 527
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The exact pathogenesis of acute lung injury (ALI) has not been fully clarified. Previous studies have demonstrated that ALI is associated with inflammation. Recent studies have demonstrated that microRNA‑421 (miR‑421) prevents inflammation in cerebellar ischemia reperfusion injury. However, the role of miR‑421 in ALI remains unclear. The present study investigated the role of miR‑421 in ALI and the mechanism underlying this. ALI was induced in vitro by treatment of RAW 264.7 macrophages with lipopolysaccharide (LPS). Cells were then transfected with miR‑421 mimic, miR‑421 mimic control, programmed cell death 4 (PDCD4) siRNA and PDCD4 siRNA control using Lipofectamine 2000. Cell viability was measured using the Cell Counting kit‑8. Reverse transcription‑quantitative polymerase chain reaction was used to detect the expression of miR‑421 and PDCD4 in RAW 264.7 macrophages. The concentrations of IL‑1β and TNF‑α were detected by ELISA. Dual‑luciferase reporter assays were used to investigate the interaction between miR‑421 and PDCD4 mRNA. LPS inhibited cell viability and miR‑421 expression. The miR‑421 mimic promoted RAW 264.7 cell viability and inhibited the expression of iNOS and COX‑2 as well as the release of IL‑1β and TNF‑α. PDCD4 siRNA inhibited the expression of iNOS and COX‑2 expression as well as the release of IL‑1β and TNF‑α. In addition, miR‑421 mimic and PDCD4 siRNA inhibit the expression of p‑NF‑κB (p65) in RAW 264.7 cells. In conclusion, the present study demonstrated the protective effect of miR‑421 on ALI and showed that miR‑421 attenuates LPS‑induced ALI by inhibiting PDCD4 and NF‑κB. These findings provided a theoretical basis for the treatment of ALI.

Introduction

Acute lung injury (ALI) is a type of severe acute lung inflammation that causes diffuse lung injury induced by various direct or indirect factors (1). The main manifestations of ALI are increased permeability of the alveolar wall and pulmonary capillaries, pulmonary interstitial and alveolar edema, eventually leading to acute respiratory insufficiency, and even acute respiratory distress syndrome (2).

The etiology and mechanism of ALI are complex. During the past half-century, there has been considerable progress in the field of ALI research, although the exact pathogenesis has not been fully clarified. Previous studies have suggested that excessive activation of inflammatory cells, release of excessive inflammatory mediators, mutual activation and the interactions of numerous inflammatory factors and effector cells, combined with uncontrolled inflammatory responses are the main pathophysiological changes (3).

A variety of inflammatory cells are recruited and activated in the lung in response to various direct and indirect harmful stimuli, leading to the release of a large number of pro-inflammatory mediators. This ‘inflammatory cascade’ of cytokines results in the induction of the inflammatory response (4). Therefore, strategies for the prevention and treatment of ALI should be focused on inhibiting the overexpression and hyperactivation of inflammatory mediators. Regulating the occurrence and development of the inflammatory response has important clinical significance in preventing the occurrence of ALI and decreasing mortality (5).

Although there are numerous strategies for ALI treatment in the clinic, including cytokine therapy, stem cell therapy and hormone therapy, the incidence rate and mortality rate of ALI remains high (6). Comprehensive elucidation of the molecular pathological mechanism of ALI is required to improve the clinical efficacy of these therapeutic strategies.

MicroRNAs (miRNAs) are small (22–25 nucleotide), non-coding RNAs that regulate gene expression by inhibiting translation or promoting the degradation of target mRNAs and regulating a variety of important cell activities by specifically pairing with the 3′-untranslated region (3′-UTR) of the target gene (7). miRNAs are also associated with the occurrence and development of lung diseases, including pneumonia, lung cancer and pulmonary fibrosis (8,9). For example, Shen et al (10) demonstrated that miR-200b may attenuate cellular senescence and inflammatory responses by targeting ZEB2 in pulmonary emphysema. Furthermore, Liang et al (11) demonstrated that miR-187 may present a novel therapeutic target in non-small-cell lung cancer based on its ability to regulate cyclins-related protein expression (11). miR-140 was also shown to suppress interstitial lung disease (ILD) development by downregulating osteoglycin (OGN) via the Wnt signaling pathway (12).

Recent studies have demonstrated the involvement of miRNAs in the regulation of the occurrence and development of the immune response that leads to ALI. For example, Huang et al (13) demonstrated that downregulation of miR-27b enhanced the expression of Nrf2 and HO-1, inhibited NF-κB signaling pathway, and protected against LPS-induced ALI in mice. In addition, Ju et al (14) reported that miR-27a attenuates LPS-induced ALI in mice by blocking activation of the TLR4/MyD88/NF-κB axis. Another study indicated that miR-326 activates the NF-κB signaling pathway by targeting the BCL2A1 gene, leading to an enhanced inflammatory response and the lung injury associated with septic shock in ALI induced in mice (15).

Recent studies have demonstrated that miR-421 regulates inflammatory responses. Zheng et al (16) reported that miR-421 inhibited p65 mRNA translation by targeting YTHDF1 to prevent inflammation in cerebral ischemia reperfusion injury. In addition, Jiang et al (17) demonstrated that miR-421 promotes the inflammatory response of fibroblast-like synoviocytes in rheumatoid arthritis by downregulating the expression of SPRY1. Furthermore, inhibition of miR-421 promotes the development of bronchopulmonary dysplasia by upregulating Fgf10 (18). However, the effect of miR-421 on ALI is not clear. In this study, we used the lipopolysaccharide-induced model of ALI to investigate the effect of miR-421 on ALI in vitro and the underlying mechanism.

Materials and methods

RAW 264.7 macrophages cells culture

RAW 264.7 macrophage cells were purchased from the American Type Culture Collection (ATCC). RAW 264.7 cells were cultured in Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% (volume/volume) fetal bovine serum (FBS; HyClone), 100 U penicillin and 100 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C under 5% CO2 in a humidified incubator. For the LPS-induced model of ALI, RAW 264.7 cells were plated into 12-well plates (1×106 cells/well) and incubated for 24 h before treatment with 0, 50, 100 or 200 ng/ml LPS (Beijing Solarbio Science & Technology Co., Ltd.) for a further 24 h, as previously described (19).

Transfection

miR-421 mimic, miR-421 mimic control, programmed cell death 4 (PDCD4) siRNA and PDCD4 siRNA control were designed and synthesized by GenePharma (Shanghai GenePharma Co., Ltd.). RAW 264.7 cells in the logarithmic growth phase were seeded into 6-well plates and incubated for 12 h at 37°C under 5% CO2 in a humidified incubator prior to transfection with miR-421 mimic (5′-AUCAACAGACAUUAAUUGGGCGC-3′, 20 µM), miR-421 mimic control (5′-UUCUCCGAACGUGUCACGUTT-3′, 20 µM), PDCD4 siRNA (5′-GAGGCUAUGAGAGAAUUUATT-3′, 20 µM) and PDCD4 siRNA control (5′-UAGCCUAGUCCAAAGCAGCAT-3′, 20 µM) using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 48 h. After 48 h, RNA and protein were extracted for further experiments.

Cell Counting kit-8 (CCK-8)

Cell viability was measured using the CCK-8 (Dojindo Molecular Technologies, Inc.) method. In brief, RAW 264.7 macrophages were seeded into 96-well plates (5×103 cells/well) and incubated for 12 h at 37°C under 5% CO2 in a humidified incubator. Subsequently, 10 µl CCK-8 reagent was added to each well and the RAW 264.7 cells were incubated for 1 h. The optical density in each well was measured at 450 nm using an iMark Microplate Absorbance Reader (Bio-Rad Laboratories, Inc.).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RAW 264.7 cells were harvested and total RNA was extracted with RNAiso (Takara Bio, Inc.) according to manufacturer's protocols. The concentration and purity of the isolated RNA was determined using a NanoDrop spectrophotometer (Thermo Fisher Scientific, Inc.). RNA was reverse-transcribed into cDNA using PrimeScript™ RT reagent kit (Takara Bio, Inc.) and Mir-X miRNA RT-qPCR TB Green® Kit (Takara Bio, Inc.). RT was performed at 37°C for 15 min, then 85°C for 5 sec. qPCR was performed on an ABI Prism 7500 (Thermo Fisher Scientific, Inc.) using TB Green® Premix Ex Taq™ II (Tli RNaseH Plus; Takara Bio, Inc.) according to manufacturer's protocols. The thermocycling conditions were as follows: Initial denaturation at 95°C for 30 sec; 40 cycles of 95°C for 5 sec and 60°C for 31 sec. The sequences of the primers as follows: miR-21 forward, 5′-TAGCTTATCAGACTGATGTTGA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′; PDCD4 forward, 5′-AGGCCGAGGTGGGCGGATCACTTGA-3′ and reverse, 5′-GCCACCATGCCTGGCTACT-3′; and GAPDH forward, 5′-CCTCTGACTTCAACAGCGACAC-3′ and reverse, 5′-TGGTCCAGGGGTCTTACTCC-3′. The relative expression level was then calculated using 2−ΔΔCq method (20).

Western blotting

Total proteins were extracted from RAW 264.7 cells using a RIPA kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocols. Nuclear proteins were extracted using the Nuclear and Cytoplasmic Protein Extraction kit (Beyotime Institute of Biotechnology). The protein concentration was measured with BCA kits (Beyotime Institute of Biotechnology) according to the manufacturer's protocols. The proteins were separated by 10% SDS-PAGE for 1.5 h. Next, the protein was transfected into PVDF (Millipore) membrane (40 µg/lane). The PVDF membrane was blocked with 5% skimmed milk at room temperature for 1 h before incubation overnight at 4°C with primary antibodies for the detection of p-NF-κB (p65) (cat. no. ab76302; dilution, 1:1,000; Abcam), NF-κB (p65; cat. no. ab76311; dilution, 1:1,000; Abcam) and β-actin (cat. no. ab8227; dilution, 1:2,000; Abcam). Membranes were then incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit secondary antibodies (cat. no. ab6721; 1:5,000; Abcam) for 1 h at room temperature. Immunoreactive bands were visualized using the ECL Plus Western Blotting Substrate (Pierce; Thermo Fisher Scientific, Inc.) kit and the FluorChem FC3 imaging system (ProteinSimple).

ELISA

RAW 264.7 cells in the logarithmic growth phase were seeded into 6-well plates and cultured at 37°C under 5% CO2 in a humidified incubator for 24 h. The culture supernatant was collected and the concentrations of IL-1β (cat. no. MLB00C) and TNF-α (cat. no. MTA00B) were detected using ELISA kits (R&D Systems, Inc.) according to the manufacturer's protocols. The optical density at 450 nm was measured using an iMark Microplate Absorbance Reader (Bio-Rad Laboratories, Inc.).

Dual-luciferase reporter assay

StarBase 3.0 (starbase.sysu.edu.cn/) was used to predict that miR-421 binds to the 3′-UTR of PDCD4 mRNA. PDCD4 reporter plasmids (pmirGLO) with the wild-type miR-421 binding site (pmirGLO-PDCD4-WT) and a mutant-type miR-421 binding site (pmirGLO-PDCD4-MUT) in the 3′-UTR of PDCD4 mRNA were synthesized by Shanghai GenePharma Co., Ltd. using a site-directed mutagenesis kit (cat. no. 200518; Agilent Technologies Inc.). Next, pmirGLO-PDCD4-WT and pmirGLO-PDCD4-MUT were co-transfected with synthesized reporter vectors (0.8 µg) and miR-421 mimic (0.8 µg) or miR-421 mimic control (0.8 µg) using Lipofectamine 2000 reagent at 37°C for 48 h. Renilla luciferase activity was measured using a Dual-Luciferase Reporter Assay system (Promega Corporation), according to the manufacturer's protocol.

Statistical analysis

SPSS 19.0 (IBM Corp.) was used for statistical analysis, and the measurement data are expressed as the mean ± standard deviation. Differences between two groups were compared using t-tests. Differences between multiple groups were compared using one-way analysis of variance, followed by Tukey's post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

LPS inhibits RAW 264.7 macrophage cell viability and miR-421 expression

RAW 264.7 macrophages were treated with different concentrations of LPS for 24 h. At 100 ng/ml, LPS significantly inhibited the viability of RAW 264.7 cells. Furthermore, the inhibitory effect of LPS at 200 ng/ml was greater than that at 100 ng/ml (Fig. 1A). Therefore, LPS at 100 ng/ml was selected as the suboptimal inhibitory dose and LPS was added at 100 ng/ml in the subsequent experiments. Furthermore, RT-qPCR demonstrated that LPS (100 ng/ml) significantly inhibited miR-421 expression in RAW 264.7 cells (Fig. 1B).

miR-421 upregulation inhibits the release of inflammatory factors by RAW 264.7 macrophages

The miR-421 mimic significantly increased miR-421 expression in RAW 264.7 macrophages (Fig. 2A). LPS (100 ng/ml) treatment inhibited RAW 264.7 cell viability. In addition, the miR-421 mimic reversed the inhibitory effect of LPS (100 ng/ml) on RAW 264.7 cell viability (Fig. 2B). The miR-421 mimic inhibited the LPS-induced expression of COX-2 and iNOS in RAW 264.7 cells (Fig. 2C and D). LPS (100 ng/ml) treatment promoted the release of IL-1β and TNF-α by RAW 264.7 cells and this effect was inhibited by the miR-421 mimic (Fig. 2E).

miR-421 directly inhibits PDCD4 expression and PDCD4 downregulation inhibits RAW 264.7 cell production of inflammatory factors

Using StarBase 3.0, it was predicted that miR-421 binds to a specific site in the 3′-UTR of PDCD4 mRNA (Fig. 3A). Dual-luciferase reporter assay confirmed that miR-421 directly targeted the 3′-UTR of PDCD4 mRNA to inhibit its expression (Fig. 3B). Further studies on the function of the PDCD4 gene demonstrated that PDCD4 siRNA blocked the inhibitory effect of LPS on RAW 264.7 cell viability (Fig. 4A and B). PDCD4 siRNA also inhibited LPS-induced expression of COX-2 and iNOS in RAW 264.7 cells (Fig. 4C and D). In addition, PDCD4 siRNA inhibited the LPS-induced increase of IL-1β and TNF-α production by RAW 264.7 cells (Fig. 4E).

miR-421 mimic and PDCD4 siRNA inhibit p-NF-κB (p65) expression in RAW 264.7 cells

Western blot analysis demonstrated that the miR-421 mimic inhibited p-NF-κB (p65) expression in RAW 264.7 cells (Fig. 5A). Furthermore, PDCD4 siRNA also inhibited p-NF-κB (p65) expression in RAW 264.7 cells (Fig. 5B).

Discussion

LPS stimulates monocytes and macrophages to release pro-inflammatory factors that mediate inflammatory reactions. Excessive inflammatory reactions may cause injury to tissues such as the lung (21). LPS may also damage pulmonary vascular endothelial cells, increase vascular permeability, cause pulmonary edema and lead to progressive disorder of gas exchange. These changes are consistent with the histopathological and physiological changes of ALI. Therefore, LPS is generally considered as an ideal stimulant to induce ALI (21).

Tang et al demonstrated that increased iNOS expression serves a key role in the pathogenesis of LPS-induced ALI (22). LPS stimulates iNOS expression in a variety of cell types found in the lung, including macrophages, neutrophils and endothelial cells (23). The present study demonstrated that miR-421 alleviated the increase in iNOS expression induced in RAW 264.7 macrophages by treatment with LPS. Therefore, we hypothesized that the protective effect of miR-421 on ALI may be associated with its inhibitory effect on iNOS.

Cyclooxygenase (COX), which converts arachidonic acid into prostaglandins, is considered to serve an important role in the occurrence and development of numerous inflammatory diseases, including ALI (24). As an inducible COX subtype, COX-2 may be induced by cytokines, growth factors, viruses, LPS and other inflammatory stimuli associated with ALI (25). In the present study, COX-2 expression was significantly increased in RAW 264.7 cells following LPS stimulation, while miR-421 attenuated this change, suggesting that the protective effect of miR-421 on ALI is associated with its inhibitory effect on COX-2.

Inflammatory mediators, particularly cytokines, serve key roles in the development and prognosis of ALI (26). In the early stage of ALI, numerous inflammatory factors, including TNF-α and IL-1β, serve a decisive role in the initiation of the early inflammatory response, and may maintain the continuous development and expansion of inflammation through a variety of mechanisms (26). While inflammatory factors may remove harmful microorganisms, excessive production of inflammatory factors will cause damage to lung tissue (27).

High levels of TNF-α may also cause severe systemic inflammatory reactions, shock, vascular leakage and pulmonary edema (28,29). In the present study, cells were stimulated with 100 ng/ml LPS for 24 h, and it was found that LPS (100 ng/ml) treatment for 24 h induced a significant increase in the production of TNF-α in RAW 264.7 cells and this effect was significantly inhibited by the miR-421 mimic.

IL-1β may also activate neutrophils, which are considered to serve a very important initiating role in the initiation of the inflammatory cascade (30,31). In the present study, it was demonstrated that LPS (100 ng/ml) treatment for 24 h induced a significant increase in the production of IL-1 β in RAW 264.7 cells and this effect was significantly inhibited by the miR-421 mimic.

Using StarBase 3.0, it was predicted that miR-421 directly targets a site in the 3′-UTR of PDCD4 mRNA. As reported previously, Yang et al (32) reported that miR-421 promotes the proliferation and invasion of non-small cell lung cancer cells through targeting PDCD4. This was also confirmed in the present study using dual-luciferase reporter assays. Furthermore, Western blot analysis demonstrated that the miR-421 mimic inhibited PDCD4 in RAW 264.7 cells. Li et al (33) demonstrated that PDCD4-overexpression significantly attenuated the anti-apoptotic effect of MSC-Exo in lung cells.

In ALI, NF-κB activation enhances the transcription of numerous pro-inflammatory factors, including adhesion molecules, cytokines and chemokines, and induces the expression of enzymes, including COX-2 and iNOS (34). It was demonstrated that PDCD4 siRNA inhibited the phosphorylation of NF-κB, indicating that NF-κB serves a role in the protective effect of miR-421 on ALI.

The results of the present study demonstrated the protective effect of miR-421 on ALI. Furthermore, it was demonstrated that miR-421 may attenuate LPS-induced ALI by inhibiting PDCD4 and NF-κB. These results provided a theoretical basis for the development of strategies for the prevention and treatment of ALI.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Inner Mongolia Autonomous Region (grant no. 2018LH08039).

Availability of data and materials

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

Authors' contributions

LW designed the experiments. HL, JS, LG, LF and DC performed the experiments. HL collected and analyzed the data. All authors confirmed the authenticity of all the raw data. HL and LW wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Szabo C, Martins V and Liaudet L: Poly(ADP-Ribose) polymerase inhibition in acute lung injury. A reemerging concept. Am J Respir Cell Mol Biol. 63:571–590. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Needham DM, Colantuoni E, Mendez-Tellez PA, Dinglas VD, Sevransky JE, Dennison Himmelfarb CR, Desai SV, Shanholtz C, Brower RG and Pronovost PJ: Lung protective mechanical ventilation and two year survival in patients with acute lung injury: Prospective cohort study. BMJ. 344:e21242012. View Article : Google Scholar : PubMed/NCBI

3 

Reagan-Steiner S, Gary J, Matkovic E, Ritter JM, Shieh WJ, Martines RB, Werner AK, Lynfield R, Holzbauer S, Bullock H, et al: Pathological findings in suspected cases of e-cigarette, or vaping, product use-associated lung injury (EVALI): A case series. Lancet Respir Med. 8:1219–1232. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Do-Umehara H, Chen C, Urich D, Zhou L, Qiu J, Jang S, Zander A, Baker MA, Eilers M, Sporn PH, et al: Suppression of inflammation and acute lung injury by Miz1 via repression of C/EBP-δ. Nat Immunol. 14:461–469. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Vlaar AP and Juffermans NP: Transfusion-related acute lung injury: A clinical review. Lancet. 382:984–994. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Needham DM, Dinglas VD, Morris PE, Jackson JC, Hough CL, Mendez-Tellez PA, Wozniak AW, Colantuoni E, Ely EW, Rice TW, et al: Physical and cognitive performance of patients with acute lung injury 1 year after initial trophic versus full enteral feeding. EDEN trial follow-up. Am J Respir Crit Care Med. 188:567–576. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Witwer KW and Halushka MK: Toward the promise of microRNAs-enhancing reproducibility and rigor in microRNA research. RNA Biol. 13:1103–1116. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Rezaei S, Mahjoubin-Tehran M, Aghaee-Bakhtiari SH, Jalili A, Movahedpour A, Khan H, Moghoofei M, Shojaei Z R, Hamblin M and Mirzaei H: Autophagy-related MicroRNAs in chronic lung diseases and lung cancer. Crit Rev Oncol Hematol. 153:1030632020. View Article : Google Scholar : PubMed/NCBI

9 

Cushing L, Jiang Z, Kuang P and Lü J: The roles of microRNAs and protein components of the microRNA pathway in lung development and diseases. Am J Respir Cell Mol Biol. 52:397–408. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Shen Z, Xuan W, Wang H, Sun F, Zhang C, Gong Q and Ge S: miR-200b regulates cellular senescence and inflammatory responses by targeting ZEB2 in pulmonary emphysema. Artif Cells Nanomed Biotechnol. 48:656–663. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Liang Z, Xu J, Ma Z, Li G and Zhu W: miR-187 suppresses non-small-cell lung cancer cell proliferation by targeting FGF9. Bioengineered. 11:70–80. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Shi S and Li H: Overexpressed microRNA-140 inhibits pulmonary fibrosis in interstitial lung disease via the Wnt signaling pathway by downregulating osteoglycin. Am J Physiol Cell Physiol. 319:C895–C905. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Huang Y, Huang L, Zhu G, Pei Z and Zhang W: Downregulated microRNA-27b attenuates lipopolysaccharide-induced acute lung injury via activation of NF-E2-related factor 2 and inhibition of nuclear factor κB signaling pathway. J Cell Physiol. 234:6023–6032. 2019. View Article : Google Scholar : PubMed/NCBI

14 

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 inflammation and apoptosis through modulating TLR4/MyD88/NF-κB pathway. Cell Cycle. 17:2001–2018. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Wu CT, Huang Y, Pei ZY, Xi X and Zhu GF: MicroRNA-326 aggravates acute lung injury in septic shock by mediating the NF-κB signaling pathway. Int J Biochem Cell Biol. 101:1–11. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Zheng L, Tang X, Lu M, Sun S, Xie S, Cai J and Zan J: microRNA-421-3p prevents inflammatory response in cerebral ischemia/reperfusion injury through targeting m6A reader YTHDF1 to inhibit p65 mRNA translation. Int Immunopharmacol. 88:1069372020. View Article : Google Scholar : PubMed/NCBI

17 

Jiang F, Zhou HY, Zhou LF, Wen YH, Gai HH and Wu GM: MicroRNA-421 promotes inflammatory response of fibroblast-like synoviocytes in rheumatoid arthritis by targeting SPRY1. Eur Rev Med Pharmacol Sci. 23:8186–8193. 2019.PubMed/NCBI

18 

Yuan HS, Xiong DQ, Huang F, Cui J and Luo H: MicroRNA-421 inhibition alleviates bronchopulmonary dysplasia in a mouse model via targeting Fgf10. J Cell Biochem. 120:16876–16887. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Lee MR, Kim JE, Park JJ, Choi JY, Song BR, Choi YW, Kim DS, Kim KM, Song HK and Hwang DY: Protective role of fermented mulberry leave extract in LPS-induced inflammation and autophagy of RAW264.7 macrophage cells. Mol Med Rep. 22:4685–4695. 2020. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Wu D, Zhang H, Wu Q, Li F, Wang Y, Liu S and Wang J: Sestrin 2 protects against LPS-induced acute lung injury by inducing mitophagy in alveolar macrophages. Life Sci. 267:1189412020. View Article : Google Scholar : PubMed/NCBI

22 

Tang L, Gao XH, Zhao B, Luo JR, Shi XY, Ge R, Ban SR and Li QS: Design and synthesis of new disubstituted benzoxazolone derivatives that act as iNOS inhibitors with potent anti-inflammatory activity against LPS-induced acute lung injury (ALI). Bioorg Med Chem. 28:1157332020. View Article : Google Scholar : PubMed/NCBI

23 

Li G, Dai Y, Tan J, Zou J, Nie X, Yang Z, Zhao J, Yang X and Chen J: SB203580 protects against inflammatory response and lung injury in a mouse model of lipopolysaccharide-induced acute lung injury. Mol Med Rep. 22:1656–1662. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Shaikh SB and Prabhakar Bhandary Y: Effect of curcumin on IL-17A mediated pulmonary AMPK kinase/cyclooxygenase-2 expressions via activation of NFκB in bleomycin-induced acute lung injury in vivo. Int Immunopharmacol. 85:1066762020. View Article : Google Scholar : PubMed/NCBI

25 

Yang HH, Duan JX, Liu SK, Xiong JB, Guan XX, Zhong WJ, Sun CC, Zhang CY, Luo XQ, Zhang YF, et al: A COX-2/sEH dual inhibitor PTUPB alleviates lipopolysaccharide-induced acute lung injury in mice by inhibiting NLRP3 inflammasome activation. Theranostics. 10:4749–4761. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Butt Y, Kurdowska A and Allen T: Acute lung injury: A clinical and molecular review. Arch Pathol Lab Med. 140:345–350. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Deng JC and Standiford TJ: Growth factors and cytokines in acute lung injury. Compr Physiol. 1:81–104. 2011.PubMed/NCBI

28 

Li X, Ye C, Mulati M, Sun L and Qian F: Ellipticine blocks synergistic effects of IL-17A and TNF-α in epithelial cells and alleviates severe acute pancreatitis-associated acute lung injury. Biochem Pharmacol. 177:1139922020. View Article : Google Scholar : PubMed/NCBI

29 

Wang X, Yan J, Xu X, Duan C, Xie Z, Su Z, Ma H, Ma H, Wei X and Du X: Puerarin prevents LPS-induced acute lung injury via inhibiting inflammatory response. Microb Pathog. 118:170–176. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Liang Y, Luo J, Yang N, Wang S, Ye M and Pan G: Activation of the IL-1β/KLF2/HSPH1 pathway promotes STAT3 phosphorylation in alveolar macrophages during LPS-induced acute lung injury. Biosci Rep. 40:BSR201935722020. View Article : Google Scholar : PubMed/NCBI

31 

Nosaka N, Martinon D, Moreira D, Crother TR, Arditi M and Shimada K: Autophagy protects against developing increased lung permeability and hypoxemia by down regulating inflammasome activity and IL-1β in LPS plus mechanical ventilation-induced acute lung injury. Front Immunol. 11:2072020. View Article : Google Scholar : PubMed/NCBI

32 

Yang YN, Bian LQ, Ling XD, Fang CY and Jiang SL: MicroRNA-421 promotes proliferation and invasion of non-small cell lung cancer cells through targeting PDCD4. Pathol Res Pract. 215:1525552019. View Article : Google Scholar : PubMed/NCBI

33 

Li JW, Wei L, Han Z and Chen Z: Mesenchymal stromal cells-derived exosomes alleviate ischemia/reperfusion injury in mouse lung by transporting anti-apoptotic miR-21-5p. Eur J Pharmacol. 852:68–76. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Al-Harbi NO, Imam F, Al-Harbi MM, Ansari MA, Zoheir KM, Korashy HM, Sayed-Ahmed MM, Attia SM, Shabanah OA and Ahmad SF: Dexamethasone attenuates LPS-induced acute lung injury through inhibition of NF-κB, COX-2, and pro-inflammatory mediators. Immunol Invest. 45:349–369. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2021
Volume 24 Issue 1

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
Liu H, Sun J, Gao L, Fan L, Chen D and Wu L: MicroRNA‑421 attenuates macrophage‑mediated inflammation by inhibiting PDCD4 <em>in vitro</em>. Mol Med Rep 24: 527, 2021
APA
Liu, H., Sun, J., Gao, L., Fan, L., Chen, D., & Wu, L. (2021). MicroRNA‑421 attenuates macrophage‑mediated inflammation by inhibiting PDCD4 <em>in vitro</em>. Molecular Medicine Reports, 24, 527. https://doi.org/10.3892/mmr.2021.12166
MLA
Liu, H., Sun, J., Gao, L., Fan, L., Chen, D., Wu, L."MicroRNA‑421 attenuates macrophage‑mediated inflammation by inhibiting PDCD4 <em>in vitro</em>". Molecular Medicine Reports 24.1 (2021): 527.
Chicago
Liu, H., Sun, J., Gao, L., Fan, L., Chen, D., Wu, L."MicroRNA‑421 attenuates macrophage‑mediated inflammation by inhibiting PDCD4 <em>in vitro</em>". Molecular Medicine Reports 24, no. 1 (2021): 527. https://doi.org/10.3892/mmr.2021.12166