miR‑625‑5p suppresses inflammatory responses by targeting AKT2 in human bronchial epithelial cells

  • Authors:
    • Fen-Hong Qian
    • Xia Deng
    • Qiong-Xin Zhuang
    • Bin Wei
    • Dan-Dan Zheng
  • View Affiliations

  • Published online on: January 3, 2019     https://doi.org/10.3892/mmr.2019.9817
  • Pages: 1951-1957
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Asthma is a common chronic inflammatory airway disease; however, whether microRNAs (miRs) could be used in the treatment of asthma remains unclear. The aim of the present study was to investigate the role of miR‑625‑5p in the inflammatory response of human bronchial epithelial cells (HBECs). Inflammation in the HBEC line, 16HBEC, was induced using different concentrations of lipopolysaccharide (LPS), which demonstrated that 1 µg/ml LPS was an appropriate concentration for further experiments. The association between protein kinase B2 (AKT2) and miR‑625‑5p was verified using a luciferase reporter assay. LPS was added to 16HBECs following the administration of miR‑625‑5p mimics or miR‑625‑5p inhibitors, and cells with silenced or overexpressed AKT2 levels. miR‑625‑5p was expressed at a high level in LPS‑activated 16HBECs. Overexpression of miR‑625‑5p inhibited interleukin (IL)‑6 and tumor necrosis factor (TNF)‑α secretion in 16HBECs. Inhibition of miR‑625‑5p enhanced LPS‑induced IL‑6 and TNF‑α secretion. miR‑625‑5p negatively regulated the expression of AKT2 in 16HBECs. A dual‑luciferase reporter assay system confirmed that miR‑625‑5p directly targeted the 3'untranslated region of AKT2. Transfection with a small interfering RNA against AKT2 inhibited inhibitor of κB phosphorylation. In brief, miR‑625‑5p may protect LPS‑induced HBECs by targeting AKT2 and inhibiting the nuclear factor‑κB signaling pathway. Therefore, miR‑625‑5p may function as an inhibitor of asthma airway inflammation in HBECs by targeting AKT2.

Introduction

Asthma is a heterogeneous disease that involves an imbalance of airway inflammation. The airway epithelium is not only the first anatomical barrier to airborne pathogens and stimuli, but it is also capable of mounting a number of immune responses (1). When an inflammatory stimulus activates epithelial cells, a series of inflammatory cascades, including innate and adaptive immune responses, are initiated (2). These important functions of airway epithelial cells make them potential targets to treat asthma.

The protein kinase B (AKT) family of kinases (AKT1, −2 and −3) has a key role in the inflammatory response. However, it is not clear which of the AKT family, if any, is a major player in asthmatic airway inflammation. In our recent experiments, it was observed that specific variants in the 3′untranslated region (UTR) of AKT2 may influence susceptibility to asthma (unpublished data); however, the details of the involvement of AKT2 activity are unknown.

MicroRNAs (miRs) are short noncoding RNAs that pair with the 3′UTR of target mRNAs to regulate gene expression through mRNA degradation or translational repression (3). An increasing body of evidence has suggested that dysregulation of the expression of miRs is correlated with various physiological and pathological processes (4), for example, in asthma (5,6). Recent studies have demonstrated the regulation of miR gene expression in bronchial epithelial cells (BECs) (69). The expression levels of miR-18A, −27A, −128, −155 and −181b-5p in asthmatic BECs decreased (6), while the level of miR-19a in the epithelia of patients with severe asthma increased (7,8). A functional study demonstrated that miR-19a inhibits the proliferation of severe asthmatic BECs by targeting transforming growth factor-β receptor 2 mRNA (9). miR-181b-5p affects eosinophilic inflammation by targeting the gene encoding secreted phosphoprotein 1 (8). For other miRs, including miR-18A, −27A, −128 and −155, no consensus miR binding sites were observed in the 3′UTRs of the target genes (5).

miR-625 is a multifunctional miR that has been implicated in carcinogenesis, including colorectal adenocarcinoma (10), malignant pleural mesothelioma (11) and hepatocellular cancer (12). To date, only one study has focused on the miR-625/AKT2 axis in increasing the chemosensitivity of glioma (13). In terms of its association with asthma, miR-625-5p is significantly downregulated in pediatric asthma and targets the gene encoding estrogen receptor 1 (14). This suggested a role for this miR in the pathogenesis of allergic diseases. Therefore, the present study was designed to investigate the novel characteristics of miR-625 in the potential pathogenic mechanism of asthma. It was hypothesized that miR-625-5p may alter the inflammatory responses in human BECs (HBECs) by targeting the AKT2 signaling pathway.

Materials and methods

Cell culture

The cell lines (16HBECs and A549 cells) were purchased from the Type Culture Collection of the Chinese Academy of Science (Shanghai, China) and grown in Dulbecco's modified Eagle's medium (DMEM; Gibco®; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% fetal bovine serum (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and 100 µg/ml streptomycin (Beyotime Institute of Biotechnology, Shanghai, China) at 37°C in 5% CO2.

MTT assay

An MTT assay (Beyotime Institute of Biotechnology, Shanghai, China) was used to measure cell viability. The cells were seeded in 96-well plates with 1–3×104 cells in a volume of 200 µl in each well and incubated for 24 h. The cells were then incubated with different concentrations (0, 0.1, 1, 10 and 100 µg/ml) of lipopolysaccharide (LPS; Sigma-Aldrich; Merck KgaA) for 12 h at 37°C. The supernatant was removed from the cells and added to medium with 5 mg/ml MTT for 4 h. The medium was discarded and 100 µl solubilization solution (included in MTT assay kit) was added to dissolve the formazan crystals. The absorbance was measured at a wavelength of 570 nm using a Multiskan Spectrum instrument (Thermo Fisher Scientific, Inc.). The ratio of the absorbance of the treatment group to that of the control group represented the viability of the cells.

Transfection

Cells seeded in 6-well plates at a density of 1×106 cells/well were transfected with an miR-625-5p mimic (5′-AGGGGGAAAGUUCUAUAGUCC-3′), miR-625-5p inhibitor (5′-AGGGGGAAAGUUCUAUAGUCC-3′), a negative control miR (5′-UUCUCCGAACGUGUCACGU-3′) and an inhibitor negative control (5′-AAGAGGCUUGCACAGUGCA-3′), which were obtained from Guangzhou Ribobio Co., Ltd., (Guangzhou, China); and with the AKT2 small interfering RNA (si-AKT2; sense, 5′-GCUCCUUCAUUGGGUACAATT-3′), scrambled negative control (5′-UUCUCCGAACGUGUCACGUTT-3′), the AKT2 overexpression plasmid (v-AKT2), and control vector, which were obtained from Shanghai GenePharma Co., Ltd., (Shanghai, China). miR-625-5p mimic (75 pmol/well) or miR-625-5p inhibitors (120 pmol/well) and their controls (corresponding concentration) were mixed with 5 µl Lipofectamine 2000™ (Thermo Fisher Scientific, Inc.), then incubated with 1×106 cells in 6-well plates. Similarly, si-AKT2 (75 pmol/well) or v-AKT2 (2.5 µg/well) were mixed with 5 µl Lipofectamine 2000™ and transfected into cells. Following 6 h, Opti-Minimum Essential Medium™ (Gibco; Thermo Fisher Scientific, Inc.) was discarded and fresh DMEM was added. The cells were incubated for another 24 h, then washed three times with cold PBS and used for protein extraction immediately after cell collection.

ELISA

The cells seeded in 6-well plates at a density of 2×105 cells/well were cultured with the miR-625-5p mimic or miR-625-5p inhibitor with LPS (1 µg/ml) for 24 h at 37°C. The cells and the culture supernatants were collected separately. The levels of interleukin (IL)-6 (cat. no. D6050) and tumor necrosis factor-α (TNF-α) (cat. no. DTA00D) in the supernatant released from 16HBECs were evaluated using ELISA kits (R&D Systems, Inc., Minneapolis, MN, USA).

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

Total RNA from cultured 16HBECs and isolated using RNAiso Plus (Takara Biotechnology Co., Ltd., Dalian, China). PrimeScript reagents (Takara Bio Inc., Otsu, Japan) were used to synthesize cDNA at 37°C for 15 min followed by RT inactivation at 85°C for 5 sec. RT-qPCR was completed using a MxPRO 3000 real-time PCR system and SYBR® Premix Ex Taq™ (TliRNaseH Plus; cat. no. RR420) (Takara Bio, Inc.); 0.8 µl primers, 0.4 µl ROX Reference Dye or Dye II 2 µl cDNA and 8 µl dH2O were employed and the reaction mixture was made up to 20 µl. The procedure was implemented according to the manufacturer's protocols. The thermocycling conditions were as follows: Denaturation at 95°C was for 5 sec, annealing at 60°C was for 34 sec min and extension at 60°C was for 1 min. The primers used are presented in Table I. The 2−∆∆Cq relative quantification method was used to calculate the mean fold expression difference between the groups (15).

Table I.

Quantitative polymerase chain reaction primers.

Table I.

Quantitative polymerase chain reaction primers.

GenePrimerSequence (5′-3′)
TNF-αForward CTCCTCACCCACACCATCA
Reverse GGAAGACCCCTCCCAGATAG
IL-6Forward TTCGGTCCAGTTGCCTTCT
Reverse GGTGAGTGGCTGTCTGTGTG
β-actinForward AGAGCTACGAGCTGCCTGAC
Reverse AGCACTGTGTTGGCGTACAG
U6Forward CTCGCTTCGGCAGCACA
Reverse AACGCTTCACGAATTTGCGT

[i] IL, interleukin; TNF, tumor necrosis factor.

Western blotting

Proteins from 16HBECs were prepared routinely using radioimmunoprecipitation lysis buffer (cat no. P0013C) and phenylmethanesulfonylfluoride (cat no: ST506) kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. A bicinchoninic assay kit (Beyotime Institute of Biotechnology) was used to quantify the protein levels. Total protein extracts (40 µg) were separated by 10% SDS-PAGE and transferred onto a polyvinylidene fluoride membrane (EMD Millipore, Billerica, MA, USA). Membranes were blocked in 5% bovine serum albumin (Beyotime Institute of Biotechnology; cat no. P0007) for 1 h at room temperature, then incubated with primary antibodies (1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA). The primary antibodies were as follows: P-AKT2 (cat. no. 8599), AKT2 (cat. no. 2964), P-IκBα (cat. no. 9246), β-actin (cat. no. 4970) overnight at 4°C, followed by incubation with horseradish peroxidase-conjugated secondary antibodies (1:3,000; cat. nos. 7076 and 7074; Cell Signaling Technology, Inc.) for 2 h at room temperature. The immunoreactive proteins on the blots were scanned by FluorChem FC3 (Protein Simple, San Jose, CA, USA).

Plasmid construction

The AKT2 3′UTR was amplified using human DNA as a template. The wild-type (WT) 3′UTR of AKT2 was inserted into the luciferase gene in the pMIR-Report vector (Promega Corporation, Madison, WI, USA; cat. no. E1330) and termed pMIR-AKT2-WT. The primers used to amplify the wild-type sequence were: 5′-CGAGCTCGGGAGGGGCCTGAAGAAGAACGA0-3′ forward, and 5′-CCAAGCTTCCTGGGCTTACTGGAGCTGCCA0-3′ reverse. pMIR-AKT2-MUT contained a mutated sequence in the 3′UTR of AKT2 inserted into the luciferase gene in the pMIR-Reporter vector. The primers used to amplify the mutant sequences were: 5′-AAGTTATATATGCGAAACCACCCAGCGGTGATGGCAGCGAG-3′ (mutated site underlined) forward; 5′-GTGGTTTCGCATATATAACTTTTTACTTAGCCTTTTTGGTT-3′ (mutated site underlined) reverse. All constructs were verified by direct sequencing by Sangon Biotech (Sangon Biotech Co. Ltd., Shanghai, China).

Dual luciferase reporter assay

The biological software Microrna (http://www.mirbase.org/) and TargetScan release 7.2 (http://www.targetscan.org/) were used to predicted as a target gene of miR-625-5p. Cells (2×104) were co-transfected in 96-well plates in triplicate with a firefly luciferase reporter plasmid (pMIR-AKT2-WT or pMIR-AKT2-MUT), a Renilla luciferase vector (pRL-SV40; Promega Corporation, Madison, WI, USA) and with miR-625-5p mimic or its control, using Lipofectamine 2000™. pRL-SV40 was used as a normalization control. Following 48 h, luciferase activity was determined using a Dual Luciferase Reporter Assay System (Promega Corporation, cat. no. E1910) according to the manufacturer's protocols and was expressed as the ratio of the firefly luciferase activity to the Renilla luciferase activity.

Statistical analysis

All experiments were repeated three times. GraphPad Prism version 5 (GraphPad Software, Inc., La Jolla, CA, USA) was used to analyze the data. The data are presented as the mean ± standard deviation for normally distributed data. A Student's t-test was used to assess the difference between two groups, while one-way analysis of variance was to analyze differences among three or more groups with a post hoc Student-Newman-Keuls test. P<0.05 was considered to indicate a statistically significance difference.

Results

Effect of LPS on the viability of 16HBECs

An MTT assay was performed to determine whether LPS influenced the viability of 16HBECs following 24 h of treatment with the various LPS concentrations (0, 0.1, 1, 10 and 100 µg/ml). LPS demonstrated a dose-dependent cytotoxic effect. A total of 1 µg/ml LPS was chosen as the best stimulatory concentration for further experiments (Fig. 1A). As presented in Fig. 1B, 16HBECs treated with LPS at 1 µg/ml significantly stimulated the cells' ability to secrete inflammatory cytokines compared with the control cells (IL-6 and TNF-α; P<0.05). 16HBECs expressed a significantly increased level of miR-625-5p compared with the A549 cell line following LPS induction (P<0.05; Fig. 1C); therefore, 16HBECs were selected for further analysis.

Effect of miR-625-5p on LPS-induced cytokine expression

ELISA and RT-qPCR were used to confirm the association between miR-625-5p and inflammatory cytokine secretion. miR-625-5p expression was significantly increased in cells transfected with the miR-625-5p mimic (P<0.05; Fig. 2A). miR-625-5p had no effect on the production of IL-6 and TNF-α in control 16HBECs. However, the mRNA and protein levels of IL-6 and TNF-α were significantly decreased in miR-625-5p mimic transfected 16HBECs following LPS-induction (P<0.05; Fig. 2B-E). Transfection with the miR-625-5p inhibitor significantly decreased miR-625-5p expression in 16HBECs when compared with the inhibitor control following LPS induction (P<0.05; Fig. 3A). Transfection with the miR-625-5p inhibitor resulted in the significantly increased expression of inflammatory cytokine mRNAs (IL-6 and TNF-α) when compared with the control in LPS treated cells (P<0.05; Fig. 3B and C). As presented in Fig. 3D and E, inhibition of miR-625-5p expression significantly upregulated LPS-induced IL-6 and TNF-α secretion.

miR-625-5p targets AKT2

The AKT2 gene was predicted as a target gene of miR-625-5p using the biological software Microrna and TargetScan. A miR-625-5p binding site was identified in the 3′UTR of AKT2 (Fig. 4A). Overexpression of miR-625-5p significantly reduced the luciferase activity in cells transfected with the pMIR-AKT2-WT vector when compared with cells transfected with the pMIR-AKT2-MUT vector (P<0.05; Fig. 4B). Western blotting was then conducted to assess the effect of miR-625-5p on the AKT2 protein level. In the miR-625-5p mimic transfected cells, the protein level of AKT2 was significantly decreased when compared with the vector only cells (P<0.05; Fig. 4C).

Effect of miR-625-5p on signaling pathways

AKT2 is involved in triggering the nuclear factor-κB (NF-κB) signaling pathway and induces inflammatory cytokines (16). To investigate whether miR-625-5p influenced the regulation of the phophoinositol-3-kinase (PI3K)/AKT pathway, the levels of phosphorylated AKT2 were quantified. The level of phosphorylated AKT2 pretreatment with the miR-625-5p mimic was significantly decreased compared with LPS treatment alone (P<0.05; Fig. 5A). Conversely, inhibition of miR-625-5p expression significantly increased the LPS-induced phosphorylation of AKT2 (P<0.05). The phosphorylation/degradation of inhibitor of κB (IκBα) is an essential step in the NF-κB signaling pathway (17). Phosphorylation of IκBα was assessed to determine whether the NF-κB signaling pathway is involved in the protective effect of miR-625-5p. The LPS-induced phosphorylation of IκBα was significantly suppressed by miR-625-5p overexpression and significantly increased by miR-625-5p inhibition compared with LPS treatment alone (P<0.05; Fig. 5B). To further confirm whether activation of the NF-κB pathway was dependent on the phosphorylation of AKT2, NF-κB activation (i.e., the phosphorylation of IκBα) was detected in LPS-stimulated 16HBECs pretreated for 24 h with si-AKT2 to knockdown AKT2 expression. Notably, treatment with si-AKT2 significantly inhibited the LPS-stimulated phosphorylation of IκBα (P<0.05), which demonstrated that the PI3K/AKT axis may be involved in NF-κB regulation in 16HBECs (Fig. 5C).

Discussion

In the present study, it was demonstrated that miR-625-5p inhibits the secretion of inflammatory mediators in HBECs. Furthermore, AKT2 was revealed to be a direct target gene of miR-625-5p. The results indicated that miR-625-5p suppresses the airway inflammatory response by downregulating AKT2, which inhibits the NF-κB signaling pathway. Therefore, miR-625-5p may function as an inhibitor of asthma airway inflammation by suppressing the inflammatory response in HBECs by directly targeting AKT2.

Bioinformatics analysis identified that the 3′UTR of human AKT2 paired miR-625-5p binding sites. The luciferase reporter assay verified the association between miR-625-5p and AKT2. Furthermore, the interactions between AKT2 and miR-625-5p in 16HBECs using a miR-625-5p mimic and inhibitor were demonstrated.

AKT2 is involved in insulin-mediated regulation of glucose homeostasis. However, a number of studies on AKT2 have focused on its role in tumors and demonstrated that AKT2 is essential for tumor growth, colony formation and cancer cell proliferation, including ovarian cancer (18), breast cancer (19), and hepatocellular carcinoma (20). According to these studies, an AKT isoform has a vital role in tumor growth, colony formation and cancer cell proliferation in other lung diseases, in addition to lung tumors; however, there are contradictory results concerning its functions. Previous studies have demonstrated that AKT2 is involved in tumor growth and colony formation, and inhibiting AKT2 decreased cellular motility and migration (2124). By contrast, another study revealed that AKT2 expression may be increased in lung tumors in a tobacco-associated model; however, loss of AKT2 expression did not lead to mutant K-ras-mediated lung tumors in a genetic model (25). In acute lung injury, Vergadi et al (26) reported that depletion of AKT2 kinase activity resulted in light acid-induced lung injury and protected mice from acid-induced lung injury (27).

The results of the present study demonstrated that miR-625-5p inhibits the secretion of TNF-α and IL-6 in 16HBECs. This was in agreement with other studies (6,28,29) in which the downregulation of multiple miRs, including microRNAs-18a, 21, −27a, −128 and −155 and miR-218 also resulted in the upregulation of IL-6 levels. However, a study by Jardim et al (30) confirmed that the expression of TNF-α was increased in asthmatic BECs, while the level of IL-6 was downregulated compared with the samples from healthy donors. This discrepancy may have been caused by the different research methods used in the two studies. Asthma is a complex disease with a variety of phenotypes (31) and there may be two possible explanations for the discrepancy between these studies. The majority of the patients selected by Jardim et al (30) had mild asthma, whereas the present study was only an in vitro simulation that did not involve samples of asthmatic tissue. In addition, in Jardim's study, the majority of the patients with asthma were receiving medication and this may have altered the expression of certain miRs. Further study is required to determine the underlying functions of miRs in asthmatic airway inflammation, which could lead to improved diagnostic technologies and subcategorization of asthma phenotypes, ultimately leading to targeted therapies for asthma.

The results of the present study demonstrated that miR-625-5p suppresses the inflammatory response of 16HBECs by silencing AKT2, which may be involved in regulating asthma. Similarly, another study revealed that the expression of miR-625-5p is markedly reduced in dust mite-induced pediatric asthma. Therefore, it has been speculated that miR-625-5p may exert protective effects in asthma (14). Indeed, lung tissue undergoes active differentiation of cells and miR-625 may serve a role in this process. Given the dysregulation of active inflammation in the asthmatic bronchi, it would not be surprising if the inaccurate differentiation of HBECs resulted in certain pathologies of asthma. In the present study, one miR targeting a single gene was focused on. However, a gene can be targeted by a number of miRs and, conversely, an miR can regulate a number of target genes. Therefore, it would be useful to characterize other miRs that may be associated with asthma to provide further options for targeted therapy. Knowledge of the function of miR-625 in inflammation is based on cell cultures. The authors' future work will demonstrate the association between miR-625 and AKT2 in murine models, which will increase our understanding of the role of miRs in asthma.

In conclusion, miR-625-5p may protect LPS-induced 16HBECs from inflammation by targeting AKT2 and inhibiting the NF-κB signaling pathway.

Acknowledgements

Not applicable.

Funding

The project was supported by the National Natural Science Foundation of China (grant no. 81370119), and the Zhenjiang Science & Technology Program (grant no. SH2015044).

Availability of data and materials

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

Authors' contributions

F-HQ and XD made substantial contributions to the design of the present study. XD, Q-XZ, BW and D-DZ performed the experiments. XD analyzed the data. F-HQ and XD wrote and revise the manuscript. All authors have 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 they have no competing interests.

References

1 

Schleimer RP, Kato A, Kern R, Kuperman D and Avila PC: Epithelium: At the interface of innate and adaptive immune responses. J Allergy Clin Immunol. 120:1279–1284. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Fang R, Cui Q, Sun J, Duan X, Ma X, Wang W, Cheng B, Liu Y, Hou Y and Bai G: PDK1/Akt/PDE4D axis identified as a target for asthma remedy synergistic with β2 AR agonists by a natural agent arctigenin. Allergy. 70:1622–1632. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Meister G and Tuschl T: Mechanisms of gene silencing by double-stranded RNA. Nature. 431:343–349. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Solberg OD, Ostrin EJ, Love MI, Peng JC, Bhakta NR, Hou L, Nguyen C, Solon M, Nguyen C, Barczak AJ, et al: Airway epithelial miRNA expression is altered in asthma. Am J RespirCrit Care Med. 186:965–974. 2012. View Article : Google Scholar

6 

Martinez-Nunez RT, Bondanese VP, Louafi F, Francisco-Garcia AS, Rupani H, Bedke N, Holgate S, Howarth PH, Davies DE and Sanchez-Elsner T: A microRNA network dysregulated in asthma controls IL-6 production in bronchial epithelial cells. PLoS One. 9:e1116592014. View Article : Google Scholar : PubMed/NCBI

7 

Matsukura S, Osakabe Y, Sekiguchi A, Inoue D, Kakiuchi Y, Funaki T, Yamazaki Y, Takayasu H, Tateno H, Kato E, et al: Overexpression of microRNA-155 suppresses chemokine expression induced by Interleukin-13 in BEAS-2B human bronchial epithelial cells. Allergol Int. 65 (Suppl):S17–S23. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Huo X, Zhang K, Yi L, Mo Y, Liang Y, Zhao J, Zhang Z, Xu Y and Zhen G: Decreased epithelial and plasma miR-181b-5p expression associates with airway eosinophilic inflammation in asthma. Clin Exp Allergy. 46:1281–1290. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Haj-Salem I, Fakhfakh R, Bérubé JC, Jacques E, Plante S, Simard MJ, Bossé Y and Chakir J: MicroRNA-19a enhances proliferation of bronchial epithelial cells by targeting TGFβR2 gene in severe asthma. Allergy. 70:212–219. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Rasmussen MH, Lyskjær I, Jersie-Christensen RR, Tarpgaard LS, Primdal-Bengtson B, Nielsen MM, Pedersen JS, Hansen TP, Hansen F, Olsen JV, et al: miR-625-3p regulates oxaliplatin resistance by targeting MAP2K6-p38 signalling in human colorectal adenocarcinoma cells. Nat Commun. 7:124362016. View Article : Google Scholar : PubMed/NCBI

11 

Kirschner MB, Cheng YY, Badrian B, Kao SC, Creaney J, Edelman JJ, Armstrong NJ, Vallely MP, Musk AW, Robinson BW, et al: Increased circulating miR-625-3p: A potential biomarker for patients with malignant pleural mesothelioma. J Thorac Oncol. 7:1184–1191. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Zhou X, Zhang CZ, Lu SX, Chen GG, Li LZ, Liu LL, Yi C, Fu J, Hu W, Wen JM and Yun JP: miR-625 suppresses tumour migration and invasion by targeting IGF2BP1 in hepatocellular carcinoma. Oncogene. 34:965–977. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Zhang J, Zhang J, Zhang J, Qiu W, Xu S, Yu Q, Liu C, Wang Y, Lu A, Zhang J and Lu X: MicroRNA-625 inhibits the proliferation and increases the chemosensitivity of glioma by directly targeting AKT2. Am J Cancer Res. 7:1835–1849. 2017.PubMed/NCBI

14 

Dong X, Xu M, Ren Z, Gu J, Lu M, Lu Q and Zhong N: Regulation of CBL and ESR1 expression by microRNA-22-3p, 513a-5p and 625-5p may impact the pathogenesis of dust mite-induced pediatric asthma. Int J Mol Med. 38:446–456. 2016. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Ozes ON, Mayo LD, Gustin JA, Pfeffer SR, Pfeffer LM and Donner DB: NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature. 401:82–85. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Sizemore N, Lerner N, Dombrowski N, Sakurai H and Stark GR: Distinct roles of the Ikappa B kinase alpha and beta subunits in liberating nuclear factor kappa B (NF-kappa B) from Ikappa B and in phosphorylating the p65 subunit of NF-kappa B. J Biol Chem. 277:3863–3869. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Linnerth-Petrik NM, Santry LA, Moorehead R, Jücker M, Wootton SK and Petrik J: Akt isoform specific effects in ovarian cancer progression. Oncotarget. 7:74820–74833. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Riggio M, Perrone MC, Polo ML, Rodriguez MJ, May M, Abba M, Lanari C and Novaro V: AKT1 and AKT2 isoforms play distinct roles during breast cancer progression through the regulation of specific downstream proteins. Sci Rep. 7:442442017. View Article : Google Scholar : PubMed/NCBI

20 

Wang Q, Yu WN, Chen X, Peng XD, Jeon SM, Birnbaum MJ, Guzman G and Hay N: Spontaneous hepatocellular carcinoma after the combined deletion of Akt isoforms. Cancer Cell. 29:523–535. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Attoub S, Arafat K, Hammadi NK, Mester J and Gaben AM: Akt2 knock-down reveals its contribution to human lung cancer cell proliferation, growth, motility, invasion and endothelial cell tube formation. Sci Rep. 5:127592015. View Article : Google Scholar : PubMed/NCBI

22 

Kim CK, Nguyen TL, Lee SB, Park SB, Lee KH, Cho SW and Ahn JY: Akt2 and nucleophosmin/B23 function as an oncogenic unit in human lung cancer cells. Exp Cell Res. 317:966–975. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Lee MW, Kim DS, Lee JH, Lee BS, Lee SH, Jung HL, Sung KW, Kim HT, Yoo KH and Koo HH: Roles of AKT1 and AKT2 in non-small cell lung cancer cell survival, growth, and migration. Cancer Sci. 102:1822–1828. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Linnerth-Petrik NM, Santry LA, Petrik JJ and Wootton SK: Opposing functions of akt isoforms in lung tumor initiation and progression. PLoS One. 9:e945952014. View Article : Google Scholar : PubMed/NCBI

25 

Hollander MC, Maier CR, Hobbs EA, Ashmore AR, Linnoila RI and Dennis PA: Akt1 deletion prevents lung tumorigenesis by mutant K-ras. Oncogene. 30:1812–1821. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Vergadi E, Vaporidi K, Theodorakis EE, Doxaki C, Lagoudaki E, Ieronymaki E, Alexaki VI, Helms M, Kondili E, Soennichsen B, et al: Akt2 deficiency protects from acute lung injury via alternative macrophage activation and miR-146a induction in mice. J Immunol. 192:394–406. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Gauna AE and Cha S: Akt2 deficiency as a therapeutic strategy protects against acute lung injury. Immunotherapy. 6:377–380. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Xu H, Sun Q, Lu L, Luo F, Zhou L, Liu J, Cao L, Wang Q, Xue J, Yang Q, et al: MicroRNA-218 acts by repressing TNFR1-mediated activation of NF-κB, which is involved in MUC5AC hyper-production and inflammation in smoking-induced bronchiolitis of COPD. Toxicol Lett. 280:171–180. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Luo F, Xu Y, Ling M, Zhao Y, Xu W, Liang X, Jiang R, Wang B, Bian Q and Liu Q: Arsenite evokes IL-6 secretion, autocrine regulation of STAT3 signaling, and miR-21 expression, processes involved in the EMT and malignant transformation of human bronchial epithelial cells. Toxicol Appl Pharmacol. 273:27–34. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Jardim MJ, Dailey L, Silbajoris R and Diaz-Sanchez D: Distinct microRNA expression in human airway cells of asthmatic donors identifies a novel asthma-associated gene. Am J Respir Cell Mol Biol. 47:536–542. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Gauthier M, Ray A and Wenzel SE: Evolving concepts of asthma. Am J Respir Crit Care Med. 192:660–668. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 19 Issue 3

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
Qian F, Deng X, Zhuang Q, Wei B and Zheng D: miR‑625‑5p suppresses inflammatory responses by targeting AKT2 in human bronchial epithelial cells. Mol Med Rep 19: 1951-1957, 2019
APA
Qian, F., Deng, X., Zhuang, Q., Wei, B., & Zheng, D. (2019). miR‑625‑5p suppresses inflammatory responses by targeting AKT2 in human bronchial epithelial cells. Molecular Medicine Reports, 19, 1951-1957. https://doi.org/10.3892/mmr.2019.9817
MLA
Qian, F., Deng, X., Zhuang, Q., Wei, B., Zheng, D."miR‑625‑5p suppresses inflammatory responses by targeting AKT2 in human bronchial epithelial cells". Molecular Medicine Reports 19.3 (2019): 1951-1957.
Chicago
Qian, F., Deng, X., Zhuang, Q., Wei, B., Zheng, D."miR‑625‑5p suppresses inflammatory responses by targeting AKT2 in human bronchial epithelial cells". Molecular Medicine Reports 19, no. 3 (2019): 1951-1957. https://doi.org/10.3892/mmr.2019.9817