eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
1/2017
vol. 42
 
Share:
Share:
Experimental immunology

MicroRNA-20b promotes the accumulation of CD11b+Ly6G+Ly6Clow myeloid-derived suppressor cells in asthmatic mice

Hua Ma
,
Shujun Guo
,
Yulan Luo
,
Yimeng Wang
,
Helong Wang
,
Jing He
,
Jie Tang
,
Lin Shen
,
Chuanwang Song

(Cent Eur J Immunol 2017; 42 (1): 30-38)
Online publish date: 2017/05/08
Article file
- MicroRNA.pdf  [0.37 MB]
Get citation
 
PlumX metrics:
 

Introduction

Bronchial asthma (here referred to as asthma) is chronic airway inflammation caused by many kinds of cells and inflammatory factors. Airway hyper-responsiveness, chronic inflammation, and airway remodeling are characteristic features of asthma [1, 2]. Epidemiological studies have found that the incidence and mortality of asthma are increasing year by year, but its pathogenesis is still unclear at present [3]. MicroRNAs (miRNAs) are non-encoding RNAs which are about 22 nucleotides in length. They regulate the expression of target genes by inhibiting their translation or causing mRNA degradation [4]. An increasing number of studies has found that many miRNAs may participate in asthma pathogenesis [5]. In an asthmatic mouse model, the upregulation of miR-221 and miR-485-3p may play an important role in the pathogenesis of asthma by downregulating the expression of Spre-2 [6]. Collison et al. demonstrated that the expression of miR-126 was upregulated in the airway wall tissues of asthmatic mice, and blocking miR-126 significantly reduced airway eosinophil cell infiltration [7]. Panganiban et al. showed that the expression of miR-1248 was upregulated in the serum of asthmatic patients, and it positively regulates the expression of interleukin (IL)-5, which is a key factor for chemotaxis, survival, and maturation of eosinophils [8]. In severe asthma, blood CD8+ T cells are activated at the same time, accompanied by reduced miR-146a/b and miR-28-5p expression [9]. In our previous study, we found that the expression of miR-20b in the lung tissues of asthmatic mice was significantly lower than that of normal mice [10]. In addition, miR-20b inhibited airway inflammation in asthmatic mice, but the mechanism remains unclear [11]. Since myeloid-derived suppressor cells (MDSCs) effectively inhibit airway inflammation in asthmatic mice [12], in this study we analyzed the effect of miR-20b on MDSCs content in the lung tissue of asthmatic mice.

Material and methods

Mice

Female BALB/c mice (2- to 3-week-old) were purchased from the Center of Experimental Animals of Bengbu Medical College. This study was approved by the Animal Care and Ethics Committee of Bengbu Medical College.

Establishment of experimental asthma model and treatment

Female BALB/c mice were randomly divided into four groups, a normal control group (Control), an asthmatic group (Asthma), a miR-20b-mimic treatment group (MiR-20b mimic) and a scrambled miR-20b treatment group (MiR-20b scrambled), with seven mice in each group. For all groups except the Control group, mice were sensitized by intraperitoneal injection with 200 µl liquid (including 50 µg of OVA and 2 mg of aluminum hydroxide) on days 0, 7, and 14. On day 21 from the start of the experiment, the mice were placed in an atomization inhalation box and were challenged with 5% OVA solution inhalation with nebulization for 30 min, continuously for 4 weeks. In addition, the MiR-20b mimic group was treated with 20 µg miR-20b mimics in 40 µl doses by nasal drip starting from the 20th day, once every three days for a total of 10 doses. The MiR-20b scrambled group received scrambled miR-20b instillation to replace the treatment with miR-20b mimics (Fig.1).

Preparation of bronchoalveolar lavage fluid (BALF)

On day 49, the mice in each group were sacrificed and tracheal intubations were performed. Mice were lavaged 6 times with 0.8 ml PBS. The BALF obtained (in total about 4.8 ml) was centrifuged at 500 × g for 5 min and the supernatant was stored at –20°C for later use in an enzyme-linked immunosorbent assay (ELISA).

Oligonucleotides, siRNAs, and primers

MiR-20b mimics, scrambled miR-20b, CREB siRNA, and CREB control siRNA were obtained from Genepharma (Shanghai, China). The miRNA sequences used were as follows: miRNA-20b mimics, sense: 5’-CAA AGU GCU CAU AGU GCA GGU AG-3’, antisense: 5’-ACC UGC ACU AUG AGC ACU UUG UU-3’; scrambled miR-20b, sense: 5’-UUC UCC GAA CGU GUC ACG UTT-3’, antisense: 5’-ACG UGA CAC GUU CGG AGA ATT-3’; CREB siRNA, sense: 5’-GUC UCC ACA AGU CCA AAC ATT-3’, antisense: 5’-UGU UUG GAC UUG UGG AGA CTT-3’; and CREB control siRNA, sense: 5’ -UUC UCC GAA CGU GUC ACG UTT-3’, antisense: 5’ -ACG UGA CAC GUU CGG AGA ATT-3’. PCR forward and reverse primers were synthesized by Sangon Biotech (Shanghai, China). The primer sequences used were: GAPDH primer, sense: 5’-GGC AAA TTC AAC GGC ACA-3’, antisense: 5’-TCC ACG ACA TAC TCA GCA CC-3’; CREB primer, sense: 5’-TGC CAC ATT AGC CCA GGT A-3’, antisense: 5’-GGG AGG ACG CCA TAA CAA CT-3’; miR-20b primer, sense: 5’-ATG CCA AAG TGC TCA TAG TG-3’, antisense: 5’-GTG CAG GGT CCG AGG T-3’; miR-20b RT primer5’-GTC GTA TCC AGT GCA GGG TCC GAG GTA TTC GCA CTG GAT ACG ACC TAC CT-3’; U6 primer, sense: 5’-CTCGCT TCG GCA GCA CA -3’, antisense: 5’-AAC GCT TCA CGA ATT TGC GT -3’; and U6 RT primer5’-AAC GCT TCA CGA ATT TGC GT-3’.

Transient transfection

CREB siRNA and control siRNA (160 nM) were transfected using Lipofectamine 2000 (Invitrogen) according to the instructions of the manufacturer. A murine blood macrophage cell line (RAW 264.7 cells; 7.2 × 105 cells/ well) was seeded into a six-well plate, and the following day, the transfected cells reached 70-90% confluence. The siRNAs and Lipofectamine 2000 were diluted with Opti-MEM with thorough mixing at room temperature for 20 min. The RAW 264.7 cells were incubated with this mixture for 48 h in standard medium. Finally, the cells were harvested for specific experiments.

Quantitative real-time PCR (qRT-PCR)

Total RNA was extracted using TRIZOL (Invitrogen). cDNA was synthesized with a TransScript First-strand cDNA Synthesis SuperMix kit from TransGen. The reaction mix included total RNA (500 ng), 1 µl anchored oligo (dT), 10 µl 2 × TS Reaction Mix, and 1 µl TransScript RT/RI Enzyme Mix (the anchored oligo (dT) was replaced with the RT primer corresponding to the target gene when the miRNA cDNA first-strand was synthesized). The reaction conditions were 30 min at 42°C then 85°C for 5 min. Quantitative PCR (qPCR) was performed with the StepOnePlus System (ABI) using the TransStartTM EcoGreen qPCR SuperMix (TransGen). The amplification conditions were 94°C for 30 s, followed by 40 cycles of 94°C for 5 s, and 60°C for 30 s. U6 RNA or GAPDH were used as endogenous controls for miRNA or other mRNA levels. The relative gene expression was analyzed using the 2¯CT method.

FACS analysis

On day 49, the lung tissues from each group were obtained. Single-cell suspensions were prepared from these lung tissues and incubated with fluorescein-conjugated antibodies for 30 min at 4°C in the dark. The stained cells were analyzed using a flow cytometer (BD FACSCaliber).

ELISA

The amount of cytokines in the serum or the BALF was determined with commercial sandwich ELISA kits (CUSABIO) according to the manufacturer’s instructions.

Western blot analysis

Briefly, RAW 264.7 cells were lysed with NP-40 lysis buffer with 1 mM PMSF. Cell lysates were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes. The membranes were blocked with 5% BSA in TBST and probed with the respective antibodies. The blots were detected using enhanced chemiluminescence according to the manufacturer’s protocol (Beyotime biotechnology).

Statistical analysis

Data are expressed as mean ± standard deviation. SPSS16.0 was used to perform single-factor analysis of variance for multiple groups. A P-value < 0.05 was considered statistically significant.

Results

MiR-20b suppresses Th2 cytokine production in the BALF of asthmatic mice

To understand the mechanism by which miR-20b inhibits allergic airway inflammation, we studied the effect of miR-20b on the production of Th1/Th2-type cytokines in mouse BALF. The results are shown in Fig. 2. Compared with the normal control group, the levels of the Th2-type cytokines IL-4 and IL-13 were significantly increased in the BALF of the asthmatic mice. However, the Th1-type cytokine IFN- was not significantly decreased in the asthmatic mice. The secretion of IL-13 and IL-4 was significantly decreased after treatment with miR-20b mimics in asthmatic mice (p < 0.01). In contrast, the production of IL-10 in each group was not significantly different in this experimental system.

MiR-20b promotes the accumulation of CD11b+Ly6G+Ly6Clow MDSCs in asthmatic mice

Since MDSCs have significant immunosuppressive activity, their relationship with asthma has recently attracted attention. To discern whether the inhibitory effect of miR-20b on airway inflammation involves MDSCs, the MDSC profiles in the lung tissues of mice in each group were assayed (Fig. 3). The results showed that there was not a significant increase of MDSCs in the asthma group compared with the normal group. After treatment with miR-20b mimics, the CD11b+Ly6G+Ly6Clow MDSC ratio was significantly elevated in asthmatic mice (p < 0.05), while the increase in CD11b+Ly6C+Ly6G- MDSCs was not statistically significant. Treatment with miR-20b-scramble did not have a significant effect on the content of MDSCs in the lung tissues of asthmatic mice.

MiR-20b increases CCL2 and TGF-β in asthmatic mice

In this study, the CCL2 content in each group was detected, and the results are shown in Fig. 4A. There was not a significant difference in CCL2 concentration between the asthmatic group and the normal group, while CCL2 levels in BALF were significantly higher after asthmatic mice treatment with miR-20b mimics. Therefore, miR-20b induced the accumulation of MDSCs accompanied by an increase in CCL2 concentration in the lungs of asthmatic mice. Because TGF- is an important effector molecule of MDSCs, the concentration of TGF- in the BALF and serum of each group of mice was assayed (Fig. 4B, C). As expected, relative to the other three groups, the concentration of TGF-, whether in the serum or in the BALF, was increased in the miR-20b-mimic treatment group.

CREB regulates miR-20b expression

At present, there have been few studies on the regulation of miR-20b expression. We used two bioinformatics software packages, P-MATCH 1.0 (http://www.gene-regulation.com/cgi-bin/pub/programs/pmatch/bin/p-match.cgi) and AliBaba2 (http://www.gene-regulation.com/pub/programs/alibaba2/index.html), to predict the transcription factors that might be involved in the regulation of miR-20b expression, in which the CREB score was higher. Thus, we attempted to verify whether CREB regulates the expression of miR-20b. The expression of miR-20b in RAW 264.7 cells transfected with CREB siRNA was analyzed by qRT-PCR analysis (Fig. 5A, B). CREB siRNA decreased the miR-20b level to 0.375-fold of the control (p < 0.01). To confirm the activity of CREB siRNA, we examined the mRNA and protein level of CREB in RAW 264.7 cells after transfection. The level of CREB mRNA decreased by 66% after CREB siRNA transfection for 48 h (Fig. 5C). The expression of the CREB protein was also decreased by 75% (Fig. 5D). These results indicate that the expression of miR-20b is regulated by CREB upstream in RAW 264.7 cells.

Discussion

Myeloid-derived suppressor cells are a group of myeloid precursor cells which fail to differentiate into mature immune cells. Having prominent immunosuppressive activity, MDSCs can significantly inhibit the function of a variety of T cells [13, 14]. Myeloid-derived suppressor cells can be divided into different subsets based on their surface molecules. In humans, MDSCs can be subdivided into the CD11b+CD33+HLA-DRCD14+ and CD11b+CD15+HLA-DRCD14 subgroups. In mice, MDSCs can be further subdivided into CD11b+Ly6G+Ly6Clow and CD11b+Ly6C+Ly6G subsets according to Ly-6C/G expression. The former group exhibits a granulocytic phenotype (G-MDSC), while the latter displays a monocytic phenotype (M-MDSC) [15, 16].
In this study, the proportion of CD11b+Ly6G+Ly6Clow MDSCs increased in lung tissue from asthmatic mice after administration of nasal drops containing miR-20b mimics, but the ratio of CD11b+Ly6C+Ly6G MDSCs was not changed. The latest research shows that many miRNAs are related to the biological functions of MDSCs, such as chemotaxis, aggregation, and functional activities [17]. Liu et al. demonstrated that miR-494 may activate the Akt pathway by targeting PTEN to regulate the accumulation and functional activity of MDSCs. In addition, miR-494 mediated the chemotaxis of MDSCs by CXCR4 chemokines in tumor tissues [18]. Li et al. showed that miR-155 and miR-21 increased GM-CSF and IL-6-induced bone marrow cell-derived MDSCs, which includes both G-MDSC and M-MDSC types. Furthermore, miR-155 and miR-21 activated STAT3 by targeting SHTP-1 and PTEN to synergistically induce the generation of MDSCs [19]. Mei et al. demonstrated that miR-200c contributed to the expansion and immunosuppressive activity of MDSCs in a mouse lung cancer and melanoma model [20]. Our previous study indicated that CCL2 signaling mediates the migration of MDSCs into the lung. In this study, we found that miR-20b can induce an increase in CCL2 concentration in the lungs of asthmatic mice [12]. Therefore, miR-20b may induce the accumulation of MDSCs by upregulating CCL2 chemokines in the lungs of asthmatic mice.
Once activated by inflammation- or tumor-driven factors, MDSCs can release a variety of biologically active mediators, including inducible nitric oxide synthase (iNOS), arginase 1, reactive oxygen species (ROS), IL-10, and transforming growth factor  (TGF-) to inhibit the function of immune cells [21, 22]. Our preliminary results indicate that tumor-derived MDSCs inhibit the allergic Th2 response in a TGF--dependent manner. In this study, we also found that the elevation of MDSCs induced by miR-20b was accompanied by an increase in TGF- in BALF and blood. Thus, miR-20b may inhibit airway inflammation in a TGF--dependent manner by inducing MDSCs in asthmatic mice.
Th1/Th2 cytokines play a key role in regulating airway inflammation in asthmatic mice [23]. It is noteworthy that the administration of miR-20b suppressed Th2 cytokines in the BALF of asthmatic mice in the current study. Recent studies have shown that miRNAs play an important role in regulating the balance of the Th1/Th2 immune response [24]. Lu et al. showed that lung eosinophil infiltration was decreased and the Th1-type cytokine IFN- was significantly increased when miR-21 was blocked in a murine asthma model. Meanwhile, DC with miR-21 deficiency increased the production of IL-12 after stimulation with LPS. miR-21-deficient mice had significantly increased Th1 delayed-type hypersensitivity [25]. These studies emphasize that miR-21 mainly regulates Th1 response rather than the Th2 reaction. Meanwhile, Mohnle et al. showed that miR-146 phosphorylated STAT4 by targeting PKC, thus resulting in Th1-cell differentiation [26]. However, our study showed that miR-20b inhibited the Th2-type response in asthmatic mice, but had no effect on Th1-type response. A study by Mattes et al. also showed that the function of Th2 cells was inhibited, and the symptoms of asthma were reduced, in mice with miR-126 ablation, and blocking miR-126 may lead to the increased expression of POU domain class 2 associating factor 1, which can negatively regulate the expression of the Th2-specific regulatory factor GATA3 [27].
This study also demonstrated that the transcription factor CREB can regulate the expression of miR-20b at the upstream level. Since the pathogenesis of asthma is closely related to miRNAs, research on the regulation of miRNAs may shed light on new asthma treatments [28]. At present, research on the regulation of miRNA expression is relatively rare. Recent studies show that the CCCTC-binding factor (CTCF) can not only control the expression of miR-125b1 and miR-375, but can also adjust the level of the miR-290 cluster in tumor cells [29]. Chavali et al. demonstrated that binding of the HMGI/Y protein to matrix attachment regions (MARs) induced histone acetylation, resulting in the expression of the miR-17-92 cluster and individual miRNAs (miR-221, 93, 17, and let-7b) in neuroblastoma cells but not in fibroblasts [30]. Zhang et al. showed that two members of the E-2b family (PEA3 and ELK-1) regulated the expression of miR-200b. Furthermore, ELK-1 inhibited the expression of miR-200b, and PEA3 promoted the expression of miR-200b [31]. Since CREB can regulate the expression of miR-20b, regulating CREB upstream of miR-20b may provide a therapeutic target for asthma treatment.
In summary, we show an important role for miR-20b in the pathogenesis of asthma, inducing the accumulation of CD11b+Ly6G+Ly6Clow cells in lung tissues. This may be a mechanism by which miR-20b suppresses airway inflammation in asthmatic mice. This study provides an experimental basis for the future clinical therapy of asthma using miR-20b as a target point of treatment.

This work was supported by National Science Foundation of China (No. 81273273) and Anhui Provincial Natural Science Foundation (1308085MH114).
The authors declare no conflict of interest.

References

1. Pelaia G, Vatrella A, Busceti MT, et al. (2015): Cellular mechanisms underlying eosinophilic and neutrophilic airway inflammation in asthma. Mediators Inflamm 2015: 879783.
2. Fahy JV (2015): Type 2 inflammation in asthma – present in most, absent in many. Nat Rev Immunol 15: 57-65.
3. Olin JT, Wechsler ME (2014): Asthma: pathogenesis and novel drugs for treatment. BMJ 349: g5517.
4. Su Z, Yang Z, Xu Y, et al. (2015): MicroRNAs in apoptosis, autophagy and necroptosis. Oncotarget 11: 8474-8490.
5. Perry MM, Adcock IM, Chung KF (2015): Role of microRNAs in allergic asthma: present and future. Curr Opin Allergy Clin Immunol 2: 156-162.
6. Liu F, Qin HB, Xu B, et al. (2015): Profiling of miRNAs in pediatric asthma: upregulation of miRNA-221 and miRNA-485-3p. Mol Med Rep 5:1178-1182.
7. Collison A, Herbert C, Siegle JS, et al. (2011): Altered expression of microRNA in the airway wall in chronic asthma: miR-126 as a potential therapeutic target. BMC Pulm Med 11: 29.
8. Panganiban RP, Pinkerton MH, Maru SY, et al. (2012): Differential microRNA epression in asthma and the role of miR-1248 in regulation of IL-5. Am J Clin Exp Immunol 2: 154-165.
9. Tsitsiou E, Williams AE, Moschos SA, et al. (2012): Transcriptome analysis shows activation of circulating CD8+ T cells in patients with severe asthma. J Allergy Clin Immunol 129: 95-103.
10. Song C, Ma H, Yao C, et al. (2012): Alveolar macrophage-derived vascular endothelial growth factor contributes to allergic airway inflammation in a mouse asthma model. Scand J Immunol 75: 599-605.
11. Ma H, Luo YL, Guo SJ, et al. (2015): Inhibitory effect of miR-20b on airway inflammation in asthmatic mice. Nan Fang Yi Ke Da Xue Xue Bao 35: 1463-1466.
12. Song C, Yuan Y, Wang XM, et al. (2014): Passive transfer of tumour-derived MDSCs inhibits asthma-related airway inflammation. Scand J Immunol 79: 98-104.
13. Nagaraj S, Youn JI, Gabrilovich DI (2013): Reciprocal relationship between myeloid-derived suppressor cells and T cells. J Immunol 191: 17-23.
14. Lindau D, Gielen P, Kroesen M, et al. (2013): The immunosuppressive tumour network: myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology 138: 105-115
15. Gabrilovich DI, Nagaraj S (2009): Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9: 162-174.
16. Jiang J, Guo W, Liang X (2014): Phenotypes, accumulation, and functions of myeloid-derived suppressor cells and associated treatment strategies in cancer patients. Hum Immunol 75: 1128-1137.
17. Chen S, Zhang Y, Kuzel TM, et al. (2015): Regulating Tumor Myeloid-Derived Suppressor Cells by MicroRNAs. Cancer Cell Microenviron 2 pii: e637.
18. Liu Y, Lai L, Chen Q, et al. (2012): MicroRNA-494 is required for the accumulation and functions of tumor-expanded myeloid-derived suppressorcells via targeting of PTEN. J Immunol 188: 5500-5510.
19. Li L, Zhang J, Diao W, et al. (2014): MicroRNA-155 and MicroRNA-21 promote the expansion of functional myeloid-derived suppressor cells. J Immunol 192: 1034-1043.
20. Mei S, Xin J, Liu Y, et al. (2015): MicroRNA-200c Promotes Suppressive Potential of Myeloid-Derived Suppressor Cells by Modulating PTEN andFOG2 Expression. PLoS One 10: e0135867.
21. Markowitz J, Wesolowski R, Papenfuss T, et al. (2013): Myeloid-derived suppressor cells in breast cancer. Breast Cancer Res Treat 140: 13-21.
22. Ostanin DV, Bhattacharya D (2013): Myeloid-derived suppressor cells in the inflammatory bowel diseases. Inflamm Bowel Dis 19: 2468-2477.
23. Tang F, Wang F, An L, et al. (2015): Upregulation of Tim-3 on CD4(+) T cells is associated with Th1/Th2 imbalance in patients with allergic asthma. Int J Clin Exp Med 8: 3809-3816.
24. Luo Y, Deng Y, Tao Z, et al. (2014): Regulatory effect of microRNA-135a on the Th1/Th2 imbalance in a murine model of allergic rhinitis. Exp Ther Med 8: 1105-1110.
25. Lu TX, Hartner J, Lim EJ, et al. (2011): MicroRNA-21 limits in vivo immune response-mediated activation of the IL-12/IFN-gamma pathway, Th1polarization, and the severity of delayed-type hypersensitivity. J Immunol 187: 3362-3373.
26. Möhnle P, Schütz SV, van der Heide V, et al. (2015): MicroRNA-146a controls Th1-cell differentiation of human CD4+ T lymphocytes by targeting PRKC. Eur J Immunol 45: 260-272.
27. Mattes J, Collison A, Plank M, et al. (2009): Antagonism of microRNA-126 suppresses the effector function of TH2 cells and the development of allergic airways disease. Proc Natl Acad Sci U S A 106: 18704-18709.
28. Kai W, Qian XU, Qun WU (2015): MicroRNAs and Asthma Regulation. Iran J Allergy Asthma Immunol 14: 120-125.
29. Saito Y, Saito H (2012): Role of CTCF in the regulation of microRNA expression. Front Genet 3: 186.
30. Chavali PL, Funa K, Chavali S, et al. (2011): Cis-regulation of microRNA expression by scaffold/matrix-attachment regions. Nucleic Acids Res 39: 6908-6918.
31. Zhang X, Zhang B, Gao J, et al. (2013): Regulation of the microRNA 200b (miRNA-200b) by transcriptional regulators PEA3 and ELK-1 protein affectsexpression of Pin1 protein to control anoikis. J Biol Chem 288: 32742-32752.
Copyright: © 2017 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.