MicroRNA‑20a promotes inflammation via the nuclear factor‑κB signaling pathway in pediatric pneumonia

  • Authors:
    • Zhihong Liu
    • Haiying Yu
    • Qiuye Guo
  • View Affiliations

  • Published online on: October 26, 2017     https://doi.org/10.3892/mmr.2017.7899
  • Pages: 612-617
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pneumonia is a common respiratory disease worldwide, which is preventable and treatable; however, it is recognized as a leading cause of mortality in children. The present study aimed to investigate the role and mechanism of microRNA (miR)‑20a in inflammation in pediatric pneumonia. Clinical serum samples were collected from children with pneumonia and healthy children. Initially, the serum expression levels of miR‑20a were detected by reverse transcription‑quantitative polymerase chain reaction. Subsequently, A549 cells were randomly divided into four groups: Control group; lipopolysaccharide (LPS; 1 µg/ml) group; LPS + miR‑20a group; and LPS + miR‑20a + pyrrolidine dithiocarbamate (PDTC; 100 mmol/l) group. The concentrations of interleukin‑6 (IL‑6), tumor necrosis factor (TNF)‑α and C‑reactive protein (CRP) in clinical serum samples and A549 cells were determined by ELISA. In addition, the protein expression levels of inhibitor of nuclear factor (NF)‑κB α (IκBα) and phosphorylated (p)‑NF‑κB were measured by western blotting. The results demonstrated that miR‑20a was upregulated in children with pneumonia and in lung cells with LPS‑induced inflammatory injury (P<0.01). In addition, compared with the LPS group, cells in the LPS + miR‑20a group exhibited increased expression levels of IL‑6, TNF‑α and CRP (P<0.05). Overexpression of miR‑20a also resulted in upregulation of the expression levels of IκBα and p‑NF‑κB compared with in the LPS group (P<0.05). Furthermore, treatment with the NF‑κB inhibitor PDTC inhibited the expression of inflammatory factors compared with in the LPS + miR‑20a group (P<0.05). In conclusion, the present study indicated that miR‑20a is upregulated in pediatric pneumonia, and overexpression of miR‑20a may promote inflammation through activation of the NF‑κB signaling pathway.

Introduction

Pneumonia is a common respiratory disease worldwide, which is preventable and treatable (1); however, it is recognized as a leading cause of mortality in children (2). It has been reported that between 1.1 and 1.4 million children succumb to pneumonia annually (3). Due to poor health care or incomplete treatment, pediatric pneumonia often recurs, which can result in numerous severe complications and a poor prognosis, thereby affecting the child's development, even leading to mortality (4,5). Although the prevalence of pediatric mortality has been reduced due to recent surgical and medical advances, pediatric pneumonia remains at a high risk of recurrence and hospitalization (6,7). Therefore, it is essential to investigate the underlying pathogenesis and to identify effective therapeutic targets for the treatment of pediatric pneumonia.

MicroRNAs (miRNAs/miRs) are a group of small non-coding RNAs, between 19 and 24 nucleotides long, which regulate the expression of target genes by binding with complementary sites on target 3′-untranslated region (3′UTR) (8). miRNAs have been reported to be implicated in the majority of cellular processes, including cell proliferation, apoptosis, migration, differentiation and metabolism, as well as in innate immunity, inflammation and infection (9,10). Previous studies have demonstrated that miRNAs serve potential roles in some types of cancer, through regulating gene expression levels and cancer pathogenesis-associated pathways, including in colorectal cancer (11), chronic lymphocytic leukemia (12), breast cancer (13), hepatocellular carcinoma (14) and lung cancer (15).

In addition to the effects of miRNAs on cancer, much attention has been paid to the potential roles of miRNAs in pediatric lung development (16) and respiratory diseases (17). A previous study suggested that numerous miRNAs associated with inflammation (miR-132, miR-181a, miR-221 and miR-222) may serve a role in the pathogenesis of Streptococcus pneumoniae meningitis (18). Recently, miR-20a has been reported to be upregulated and participate in the regulation of cell proliferation in lung cancer (19). However, it is unclear whether miR-20a is associated with the pathogenesis of pediatric pneumonia.

The present study evaluated the expression levels of miR-20a in children with pneumonia and in a cell model of lipopolysaccharide (LPS)-induced inflammatory lung injury (20). Subsequently, the role and mechanisms of miR-20a in pediatric pneumonia-associated inflammation were investigated.

Materials and methods

Patients

A total of 16 children (8 male and 8 female; mean age ± standard deviation, 11.4±2.16 years; age range, 8–15) with pneumonia were recruited to the present study. Pneumonia was diagnosed according to clinical presentation, respiratory symptoms, bacterial infection and chest X-ray. In addition, 16 gender-matched children (mean age ± standard deviation, 11.3±2.24 years; age range, 7–15) with fever were included as the control group; these children were treated in the Emergency Department and had no respiratory symptoms. These 32 patients were recruited from the Jinan Maternity and Child Care Hospital (Jinan, China) between June 2012 and December 2013. None of the patients underwent any treatment prior to blood collection. Venous blood samples (5 ml) were collected from the patients and the present study was approved by the Ethics Committee of the Jinan Maternity and Child Care Hospital. Written informed consent was obtained from the legal guardians of all participants.

Cell culture and treatment

The A549 human lung adenocarcinoma cell line was purchased from the Shanghai Cell Bank of Chinese Academy of Sciences (Shanghai, China). Cells were cultured in RPMI 1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin (both Beijing Transgen Biotechnology Co., Ltd., Beijing, China) at 37°C in an incubator containing 5% CO2. A total of 2 ml cells/well, at a concentration of 5×105 cells/ml, were cultured in a 6-well plate. Subsequently, cells were separated into groups, and were treated with LPS (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), miR-20a mimic (sense 5′-UAAAGUGCUUAUAGUGCAGGUAG-3′ and antisense 5′-ACCUGCACUAUAAGCACUUUAUU-3′), mimic control (sense 5′-UGCUUAGUAUAAGAUGCAGGUAG-3′ and antisense 5′-ACGCUAUAUCUUAUAGCACACUU-3′) (both Shanghai GenePharma Co., Ltd., Shanghai, China) or the nuclear factor (NF)-κB inhibitor pyrrolidine dithiocarbamate (PDTC; Sigma-Aldrich; Merck KGaA). Briefly, the cells were divided into the following four groups: Control group (non-treated cells); LPS group, in which cells were stimulated with LPS (1 µg/ml) for 12 h at 37°C to induce inflammatory lung cell injury (20); LPS + miR-20a group, in which cells overexpressing miR-20a were treated with LPS (1 µg/ml) for 12 h at 37°C; and LPS + miR-20a + PDTC group, in which cells overexpressing miR-20a were treated with PDTC (100 mmol/l) for 30 min at 37°C and then treated with LPS (1 µg/ml) for 12 h at 37°C (21). In addition, an LPS + mimic control group, in which cells were transfected with mimic control and then treated with LPS (1 µg/ml) for 12 h at 37°C, was generated. To overexpress miR-20a, the miR-20a mimic (50 nM) was transfected into A549 cells (1×105 cells) using 20 µl Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's protocol. Transfection with the mimic control was the same as the transfection of miR-20a. Transfected cells were harvested at 48 h post-transfection for subsequent experiments.

ELISA analysis

Blood samples were collected from the children and were centrifuged at 1,500 × g for 10 min at 4°C. The supernatant serum samples were then collected and stored at −80°C. The serum concentrations of interleukin (IL)-6 (cat. no. S6050), tumor necrosis factor (TNF)-α (cat. no. STA00C) and C-reactive protein (CRP; cat. no. SCRP00) were determined using corresponding ELISA kits (R&D Systems, Inc., Minneapolis, MN, USA) according to the manufacturer's protocols. The absorbance was measured at 450 nm using a microplate reader (Thermo Fisher Scientific, Inc.). The concentrations of IL-6, TNF-α and CRP were calculated based on the standard curve.

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

Total RNA was extracted from blood samples using the RNAprep pure Blood kit (Tiangen Biotech, Beijing, China) and total RNA was extracted from treated cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Subsequently, RNA was measured using a UV spectrophotometer (BD Biosciences, San Diego, CA, USA) at A260, and cDNA was generated from miRNAs using stem-loop RT primers (Applied Biosystems; Thermo Fisher Scientific, Inc.) with the following cycling parameters: 30°C for 10 min, followed by 42°C for 30 min and 95°C for 5 min. Primer sequences for miR-20a and U6 small nuclear (sn)RNA are presented in Table I. SYBR® Premix Ex Taq kit (Takara Biotechnology Co., Ltd., Dalian, China) was used to perform PCR amplification; the PCR program was as follows: 94°C for 10 min, followed by 40 cycles at 94°C for 15 sec, 56°C for 30 sec and 72°C for 1 min, and finally 72°C for 10 min. U6 snRNA was used as an internal control. Relative quantification and calculations were conducted using the quantification cycle (Cq) method (2−ΔΔCq) (22).

Table I.

Primer sequences for specific genes.

Table I.

Primer sequences for specific genes.

GenePrimerSequence (5′-3′)
miR-20aForward GCCGCGCTAAAGTGCTTATAGTG
Reverse CACCAGGGTCCGAGGT
U6Forward TGCGGGTGCTCGCTTCGGCAGC
Reverse CCAGTGCAGGGTCCGAGGT

[i] miR-20a, microRNA-20a.

Western blotting

The cells were collected and the proteins were extracted using RIPA buffer (Beyotime Institute of Biotechnology, Shanghai, China) containing a cocktail of protease inhibitors (Roche Diagnostics, Basel, Switzerland). Bicinchoninic acid protein assay kit (Sangon Biotech Co., Ltd., Shanghai, China) was used to determine protein concentration. Subsequently, equal amounts of protein (30 µg/lane) were separated by 10% SDS-PAGE and were transferred to a polyvinylidene fluoride membrane. After blocking with 5% non-fat milk at room temperature for 2 h, the membrane was incubated with rabbit anti-human inhibitor of NF-κB α (IκBα; cat. no. SAB4501994; 1:500), phosphorylated (p)-NF-κB (cat. no. SAB4301496; 1:500) polyclonal antibodies and β-actin monoclonal antibody (cat. no. SAB2100037; 1:1,000) (all Sigma-Aldrich; Merck KGaA) at 4°C overnight. β-actin was used as an internal control. After washing with PBS, the membrane was incubated with horseradish peroxidase-conjugated goat anti-rabbit secondary antibody (cat. no. 31460; 1:10,000; Thermo Fisher Scientific, Inc.) for 2 h at room temperature. The membrane was washed again with PBS and color was developed using a 3,3′-diaminobenzidine substrate (Sangon Biotech Co., Ltd.). Image Pro Plus software (version 5.0; Media Cybernetics, Inc., Rockville, MD, USA) was used to semi-quantify the expression levels of the target proteins.

Statistical analysis

All data are presented as the mean ± standard deviation, and SPSS 16.0 statistics software (SPSS, Inc., Chicago, IL, USA) was used to analyze the data. All data were initially tested for Gaussian distribution. Subsequently, data were analyzed by unpaired two-tailed t-test (for two groups) or one-way analysis of variance followed by a Tukey post hoc test (for more than two groups). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-20a expression in patients with pneumonia and in LPS-induced cells

To explore the expression levels of miR-20a in patients with pneumonia, an RT-qPCR analysis was performed; the results demonstrated that miR-20a expression was higher in patients with pneumonia compared with in healthy controls (P<0.01; Fig. 1A). In addition, LPS was used to induce inflammatory injury in A549 cells. Compared with in the control group, the expression levels of miR-20a were increased in the LPS group (P<0.01; Fig. 1B).

Effects of miR-20a overexpression on inflammation in pneumonia

The present study investigated the association between miR-20a and the inflammatory reaction. ELISA analysis revealed that the serum concentrations of inflammatory factors, including IL-6, TNF-α and CRP, were higher in patients with pneumonia compared with in healthy controls (P<0.05; Fig. 2A). The results of an RT-qPCR analysis indicated that the expression levels of miR-20a were significantly upregulated following transfection of cells with miR-20a mimic compared with in the mimic control group (P<0.001; Fig. 2B), confirming that miR-20a was successfully overexpressed. Subsequent experiments demonstrated that the concentrations of inflammatory factors, including IL-6, TNF-α and CRP, were increased in the LPS group compared with in the control group (P<0.05; Fig. 2C-E). In addition, overexpression of miR-20a further increased the concentrations of IL-6, TNF-α and CRP compared with the LPS group (P<0.05). The levels of inflammatory cytokines in the LPS + mimic control group were not significantly different compared with the LPS group (data not shown).

Effects of miR-20a overexpression on the NF-κB signaling pathway in LPS-induced cells

To further investigate the mechanism underlying the effects of miR-20a on the induction of inflammation, NF-κB signaling pathway-associated proteins, including IκBα and p-NF-κB, were measured by western blotting. As shown in Fig. 3A-C, the protein expression levels of IκBα and p-NF-κB were increased in the LPS group compared with in the control group (P<0.05). The expression of total NF-κB was consistent with that of β-actin (data not shown). In addition, the expression levels of IκBα and p-NF-κB were further increased in the LPS + miR-20a group compared with in the LPS group (P<0.05). The expression levels of IκBα and p-NF-κB in LPS + mimic control group were not significantly different compared with the LPS group (data not shown).

Effects of the NF-κB inhibitor PDTC on miR-20a-induced inflammation in LPS-treated cells

To evaluate the role of the NF-κB signaling pathway in miR-20a-induced inflammation, the NF-κB inhibitor PDTC was used to suppress the NF-κB signaling pathway. ELISA was subsequently used to detect the expression levels of inflammatory factors. The results demonstrated that following treatment with the NF-κB inhibitor PDTC, the expression levels of IL-6, TNF-α and CRP were markedly decreased compared with in the LPS + miR-20a group (P<0.05; Fig. 4). The levels of inflammatory cytokines in the LPS + mimic control group were not significantly different compared with the LPS group (data not shown).

Discussion

miRNAs have been reported to have a role in inflammatory diseases (23). The results of the present study demonstrated that the expression levels of miR-20a were increased in children with pneumonia and in lung cells with LPS-induced inflammatory injury. Furthermore, the results indicated that miR-20a overexpression may promote the expression of inflammatory factors, including IL-6, TNF-α and CRP. miR-20a overexpression also increased the expression levels of NF-κB signaling pathway-associated proteins, including IκBα and p-NF-κB, whereas treatment with the NF-κB inhibitor PDTC was able to inhibit the enhancing effects of miR-20a on the inflammatory reaction.

It has previously been demonstrated that miRNAs exert marked regulatory effects on inflammation, and various miRNAs have been identified and considered to participate in the pathological mechanisms of inflammatory diseases (24). Bazzoni et al (25) reported that in neutrophils and macrophages, miR-9 overexpression could be induced by LPS and negatively regulated NF-κB signaling pathway-dependent inflammatory responses. miR-21 has also been identified to target tumor suppressor genes and negatively regulate the LPS-stimulated Toll-like receptor (TLR)4 pathway, which may result in activation of NF-κB and production of IL-10 (26). Downregulated miR-125b has been indicated to promote the expression of TNF-α in LPS-induced activated macrophages (27). Furthermore, miR-155 has been reported to suppress the inflammatory reaction through regulating the expression of myeloid differentiation primary response protein 88, which serves an important role in the TLR pathway (28). Taganov et al (29) revealed that miR-146 may be significantly upregulated in human monocytic cells stimulated by LPS, and miR-146 was able to inhibit the expression of target genes, including IL-1 receptor-associated kinase 1 and TNF receptor-associated factor 6, and negatively regulate TLR signaling. It was also demonstrated that miR-146a was upregulated in monocytes stimulated by inflammatory cytokines, including TNF-α and IL-1β (29). Following activation of the TLR pathway, miR-147 has been reported to be upregulated and to inhibit the excessive inflammatory responses in murine lung macrophages (30).

The present study focused on the role of miR-20a in pediatric pneumonia. miR-20a is a member of the miR-17-92 cluster, which is involved in tumorigenesis (31). Previous studies have demonstrated that miR-20a is associated with cell proliferation (32) and cell cycle progression (33) by regulating the expression of transcription factor E2F1. Fan et al (34) also reported that miR-20a overexpression was able to promote cell proliferation and invasion by inhibiting the expression of targeted amyloid precursor protein in ovarian cancer cells. Furthermore, it has been demonstrated that miR-20 serves an important role in inflammatory responses. Philippe et al (35) reported that miR-20 could effectively inhibit the production of inflammatory cytokines, including IL-6, C-X-C motif chemokine ligand 10, IL-1β and TNF-α, in LPS-activated fibroblast-like synoviocytes. Conversely, miR-20a was reported to promote macrophage inflammatory responses through modulating the expression of signal-regulatory protein α both in vitro and in vivo (36). The various effects of miR-20 on the inflammatory response may be associated with the different target genes of miR-20 in various cell types. In the present study, a high expression of miR-20a was detected in pediatric pneumonia, and the results revealed that miR-20a overexpression increased the expression levels of IL-6, TNF-α and CRP in lung cells with LPS-induced inflammatory injury. These findings indicated that miR-20a may be involved in the inflammatory response in pediatric pneumonia.

The present study also investigated the mechanism underlying the effects of miR-20a on inflammation in pediatric pneumonia. The NF-κB signaling pathway is considered closely associated with inflammation; the activation of NF-κB induces the secretion of proinflammatory cytokines, including IL-1 and TNF-α, chemokines and adhesion molecules. Previous studies have demonstrated that NF-κB is activated in numerous inflammatory disorders, including arthritis (37), glomerulonephritis (38), gastritis (39) and asthma (40). The interaction between miRNAs (miR-21, miR-146, miR-155, miR-181b and miR-301a) and NF-κB activation has been demonstrated in human disease (41). Bhaumik et al (42) demonstrated that NF-κB activity and the inflammatory pathway can be negatively regulated by miR-146a/b in breast cancer cells. Notably, the results of the present study preliminarily confirmed that the NF-κB signaling pathway was activated by miR-20a overexpression, and the proinflammatory role of miR-20a could be inhibited following treatment of LPS-induced cells with an NF-κB inhibitor. These results suggested that miR-20a may promote inflammation by activating the NF-κB signaling pathway.

In conclusion, the present study confirmed that miR-20a was upregulated in children with pediatric pneumonia and in human lung adenocarcinoma A549 cells with LPS-induced inflammatory injury. In addition, overexpression of miR-20a was able to promote inflammation, which may be associated with the NF-κB signaling pathway in pediatric pneumonia. However, further studies are required to investigate the target molecules of miR-20a in pediatric pneumonia.

References

1 

He C, Kang L, Miao L, Li Q, Liang J, Li X, Wang Y and Zhu J: Pneumonia mortality among children under 5 in China from 1996 to 2013: An analysis from national surveillance system. PLoS One. 10:e01336202015. View Article : Google Scholar : PubMed/NCBI

2 

Wardlaw T, Salama P, Johansson EW and Mason E: Pneumonia: The leading killer of children. Lancet. 368:1048–1050. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Liu L, Johnson HL, Cousens S, Perin J, Scott S, Lawn JE, Rudan I, Campbell H, Cibulskis R, Li M, et al: Global, regional, and national causes of child mortality: An updated systematic analysis for 2010 with time trends since 2000. Lancet. 379:2151–2161. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Bhutta ZA, Das JK, Walker N, Rizvi A, Campbell H, Rudan I and Black RE: Lancet Diarrhoea and Pneumonia Interventions Study Group: Interventions to address deaths from childhood pneumonia and diarrhoea equitably: What works and at what cost? Lancet. 381:1417–1429. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Leyenaar JK, Lagu T, Shieh MS, Pekow PS and Lindenauer PK: Management and outcomes of pneumonia among children with complex chronic conditions. Pediatr Infect Dis J. 33:907–911. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Plioplys AV and Kasnicka I: Nebulized tobramycin: Prevention of pneumonias in patients with severe cerebral palsy. J Pediatr Rehabil Med. 4:155–158. 2011.PubMed/NCBI

7 

Loddenkemper T, Syed TU, Ramgopal S, Gulati D, Thanaviratananich S, Kothare SV, Alshekhlee A and Koubeissi MZ: Risk factors associated with death in in-hospital pediatric convulsive status epilepticus. PLoS One. 7:e474742012. View Article : Google Scholar : PubMed/NCBI

8 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Kloosterman WP and Plasterk RH: The diverse functions of microRNAs in animal development and disease. Dev Cell. 11:441–450. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Jovanovic M and Hengartner MO: miRNAs and apoptosis: RNAs to die for. Oncogene. 25:6176–6187. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Michael MZ, O'Connor SM, van Holst Pellekaan NG, Young GP and James RJ: Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res. 1:882–891. 2003.PubMed/NCBI

12 

Calin GA, Liu CG, Sevignani C, Ferracin M, Felli N, Dumitru CD, Shimizu M, Cimmino A, Zupo S, Dono M, et al: MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proc Natl Acad Sci USA. 101:11755–11760. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, et al: MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 65:7065–7070. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T and Shimotohno K: Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene. 25:2537–2545. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T, et al: Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 9:189–198. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Khoshgoo N, Kholdebarin R, Iwasiow BM and Keijzer R: MicroRNAs and lung development. Pediatr Pulmonol. 48:317–323. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Booton R and Lindsay MA: Emerging role of MicroRNAs and long noncoding RNAs in respiratory disease. Chest. 146:193–204. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Omran A, Jagoo M, Ashhab MU, He F, Kong H, Peng J and Yin F: MicroRNAs expression changes in acute Streptococcus pneumoniae meningitis. Translational Neurosci. 5:131–136. 2014. View Article : Google Scholar

19 

Hayashita Y, Osada H, Tatematsu Y, Yamada H, Yanagisawa K, Tomida S, Yatabe Y, Kawahara K, Sekido Y and Takahashi T: A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res. 65:9628–9632. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Qi W, Li H, Cai XH, Gu JQ, Meng J, Xie HQ, Zhang JL, Chen J, Jin XG, Tang Q, et al: Lipoxin A4 activates alveolar epithelial sodium channel gamma via the microRNA-21/PTEN/AKT pathway in lipopolysaccharide-induced inflammatory lung injury. Lab Invest. 95:1258–1268. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Romacho T, Azcutia V, Vazquez-Bella M, Matesanz N, Cercas E, Nevado J, Carraro R, Rodríguez-Mañas L, Sánchez-Ferrer CF and Peiró C: Extracellular PBEF/NAMPT/visfatin activates pro-inflammatory signalling in human vascular smooth muscle cells through nicotinamide phosphoribosyltransferase activity. Diabetologia. 52:2455–2463. 2009. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Plank M, Maltby S, Mattes J and Foster P: Targeting translational control as a novel way to treat inflammatory disease: The emerging role of microRNAs. Clin Exp Allergy. 43:981–999. 2013. View Article : Google Scholar : PubMed/NCBI

24 

O'Connell RM, Rao DS and Baltimore D: microRNA regulation of inflammatory responses. Annu Rev Immunol. 30:295–312. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Bazzoni F, Rossato M, Fabbri M, Gaudiosi D, Mirolo M, Mori L, Tamassia N, Mantovani A, Cassatella MA and Locati M: Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals. Proc Natl Acad Sci USA. 106:5282–5287. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Sheedy FJ, Palsson-McDermott E, Hennessy EJ, Martin C, O'Leary JJ, Ruan Q, Johnson DS, Chen Y and O'Neill LA: Negative regulation of TLR4 via targeting of the proinflammatory tumor suppressor PDCD4 by the microRNA miR-21. Nat Immunol. 11:141–147. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Tili E, Michaille JJ, Cimino A, Costinean S, Dumitru CD, Adair B, Fabbri M, Alder H, Liu CG, Calin GA and Croce CM: Modulation of miR-155 and miR-125b levels following lipopolysaccharide/TNF-alpha stimulation and their possible roles in regulating the response to endotoxin shock. J Immunol. 179:5082–5089. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Tang B, Xiao B, Liu Z, Li N, Zhu ED, Li BS, Xie QH, Zhuang Y, Zou QM and Mao XH: Identification of MyD88 as a novel target of miR-155, involved in negative regulation of Helicobacter pylori-induced inflammation. FEBS Lett. 584:1481–1486. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Taganov KD, Boldin MP, Chang KJ and Baltimore D: NF-κB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA. 103:12481–12486. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Liu G, Friggeri A, Yang Y, Park YJ, Tsuruta Y and Abraham E: miR-147, a microRNA that is induced upon Toll-like receptor stimulation, regulates murine macrophage inflammatory responses. Proc Natl Acad Sci USA. 106:15819–15824. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Olive V, Jiang I and He L: mir-17-92, a cluster of miRNAs in the midst of the cancer network. Int J Biochem Cell Biol. 42:1348–1354. 2010. View Article : Google Scholar : PubMed/NCBI

32 

O'Donnell KA, Wentzel EA, Zeller KI, Dang CV and Mendell JT: c-Myc-regulated microRNAs modulate E2F1 expression. Nature. 435:839–843. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Pickering MT, Stadler BM and Kowalik TF: miR-17 and miR-20a temper an E2F1-induced G1 checkpoint to regulate cell cycle progression. Oncogene. 28:140–145. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Fan X, Liu Y, Jiang J, Ma Z, Wu H, Liu T, Liu M, Li X and Tang H: miR-20a promotes proliferation and invasion by targeting APP in human ovarian cancer cells. Acta Biochim Biophys Sin (Shanghai). 42:318–324. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Philippe L, Alsaleh G, Pichot A, Ostermann E, Zuber G, Frisch B, Sibilia J, Pfeffer S, Bahram S, Wachsmann D and Georgel P: MiR-20a regulates ASK1 expression and TLR4-dependent cytokine release in rheumatoid fibroblast-like synoviocytes. Ann Rheum Dis. 72:1071–1079. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Zhu D, Pan C, Li L, Bian Z, Lv Z, Shi L, Zhang J, Li D, Gu H, Zhang CY, et al: MicroRNA-17/20a/106a modulate macrophage inflammatory responses through targeting signal-regulatory protein α. J Allergy Clin Immunol. 132:426–436. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Han Z, Boyle DL, Manning AM and Firestein GS: AP-1 and NF-kB regulation in rheumatoid arthritis and murine collagen-induced arthritis. Autoimmunity. 28:197–208. 1998. View Article : Google Scholar : PubMed/NCBI

38 

Sakurai H, Hisada Y, Ueno M, Sugiura M, Kawashima K and Sugita T: Activation of transcription factor NF-κappa B in experimental glomerulonephritis in rats. Biochim Biophys Acta. 1316:132–138. 1996. View Article : Google Scholar : PubMed/NCBI

39 

van Den Brink GR, ten Kate FJ, Ponsioen CY, Rive MM, Tytgat GN, van Deventer SJ and Peppelenbosch MP: Expression and activation of NF-κappa B in the antrum of the human stomach. J Immunol. 164:3353–3359. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Hart LA, Krishnan VL, Adcock IM, Barnes PJ and Chung KF: Activation and localization of transcription factor, nuclear factor-κappaB, in asthma. Am J Respir Crit Care Med. 158:1585–1592. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Ma X, Buscaglia Becker LE, Barker JR and Li Y: MicroRNAs in NF-κB signaling. J Mol Cell Biol. 3:159–166. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Bhaumik D, Scott GK, Schokrpur S, Patil CK, Orjalo AV, Rodier F, Lithgow GJ and Campisi J: MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging (Albany NY). 1:402–401. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 17 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 Z, Yu H and Guo Q: MicroRNA‑20a promotes inflammation via the nuclear factor‑κB signaling pathway in pediatric pneumonia. Mol Med Rep 17: 612-617, 2018
APA
Liu, Z., Yu, H., & Guo, Q. (2018). MicroRNA‑20a promotes inflammation via the nuclear factor‑κB signaling pathway in pediatric pneumonia. Molecular Medicine Reports, 17, 612-617. https://doi.org/10.3892/mmr.2017.7899
MLA
Liu, Z., Yu, H., Guo, Q."MicroRNA‑20a promotes inflammation via the nuclear factor‑κB signaling pathway in pediatric pneumonia". Molecular Medicine Reports 17.1 (2018): 612-617.
Chicago
Liu, Z., Yu, H., Guo, Q."MicroRNA‑20a promotes inflammation via the nuclear factor‑κB signaling pathway in pediatric pneumonia". Molecular Medicine Reports 17, no. 1 (2018): 612-617. https://doi.org/10.3892/mmr.2017.7899