Long non‑coding RNA GAS5 protects against Mycoplasma pneumoniae pneumonia by regulating the microRNA‑222‑3p/TIMP3 axis

  • Authors:
    • Likun Yang
    • Xifeng Zhang
    • Xiufen Liu
  • View Affiliations

  • Published online on: March 17, 2021     https://doi.org/10.3892/mmr.2021.12019
  • Article Number: 380
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mycoplasma pneumoniae pneumonia (MPP) is a type of pneumonia induced by M. pneumoniae (MP) infection. The present study investigated the effect of long non‑coding RNA growth arrest‑specific 5 (GAS5) in MPP and the underlying molecular mechanism of this. The expression of GAS5, microRNA‑222‑3p, (miR‑222‑3p) and tissue inhibitor of metalloproteinases‑3 (TIMP3) in MPP was investigated using reverse transcription‑quantitative PCR. Lipid‑associated membrane protein (LAMP)‑induced THP‑1 cells were used to model MPP. The viability of LAMP‑induced THP‑1 cells was analyzed using an MTT assay. Expression levels of interleukin (IL)‑1β, IL‑6 and tumor necrosis factor‑α (TNF‑α) pro‑inflammatory cytokines, and the anti‑inflammatory cytokine heme oxygenase‑1 (HO‑1) in LAMP‑induced THP‑1 cells were measured by ELISA. A dual‑luciferase reporter assay assessed the associations among GAS5, miR‑222‑3p and TIMP3. The expression of GAS5 and TIMP3 was downregulated in MPP. Expression of miR‑222‑3p was upregulated. GAS5‑overexpression increased the viability of LAMP‑induced THP‑1 cells. GAS5 upregulation decreased the levels of IL‑1β, IL‑6, TNF‑α and HO‑1 levels in LAMP‑induced THP‑1 cells. GAS5 directly interacted with miR‑222‑3p. TIMP3 was a target of miR‑222‑3p. miR‑222‑3p upregulation or TIMP3‑knockdown reversed the promotion effect on cell viability as well as the inhibitory effect on inflammation caused by GAS5‑overexpression in LAMP‑induced THP‑1 cells. GAS5‑overexpression increased the viability and decreased the inflammation of LAMP‑induced THP‑1 cells by regulating the miR‑222‑3p/TIMP3 axis. These results demonstrated a potential therapeutic target for MPP treatment.

Introduction

Mycoplasma pneumoniae pneumonia (MPP) is a type of pneumonia induced by M. pneumoniae infection (1). MPP frequently occurs in children and infants. The clinical presentation is low-grade fever, cough and asthma-like symptoms (2). Despite the advancement in medical science, the incidence of severe or fatal MPP continues to rise (3). Severe MPP is characterized by pulmonary fibrosis, obstructive bronchiolitis and copious pleural effusion (PE), and may be life-threatening (4). Macrolide antibiotics are the first-line therapy for MPP (5). However, macrolide resistance develops frequently in children with MPP, with a rate of 87.2% reported in one previous study (6). Alternative and effective therapeutic strategies in children with MPP are required.

Long non-coding RNAs (lncRNAs) are essential for the regulation of respiratory diseases, including idiopathic pulmonary fibrosis (IPF) (7), acute lung injury (ALI) (8), and pneumonia (9). The growth arrest-specific 5 (GAS5) lncRNA is crucial in numerous inflammatory diseases. GAS5 upregulation alleviates renal fibrosis and inflammatory reactions in rats with diabetic nephropathy (10). GAS5-silencing decreases the viability and aggravates the inflammatory injury of lipopolysaccharide (LPS)-induced chondrocytes (11). Notably, GAS5 is poorly expressed in ALI, and its overexpression attenuates inflammation in ALI mice (12). However, the precise role of GAS5 in the progression of MPP remains unclear.

As biological molecules, microRNAs (miRNAs) participate in the progression of pneumonia. miR-217 contributes toward lung injury and inflammation in interstitial pneumonia (13). miR-21 is upregulated in and promotes the development of ventilator-associated pneumonia (14). miR-155 was reported to induce impaired bacterial clearance and increase mortality in patients suffering from pneumonia that developed following influenza (15). Importantly, miR-222-3p expression is enhanced in children with MPP (16). miR-222-3p is a target of GAS5 in papillary thyroid carcinoma (17). However, the specific regulatory association between GAS5 and miR-222-3p in MPP remains to be elucidated.

The regulatory functions of miRNAs involve the targeting of mRNAs through complementary sequences (18). Tissue inhibitor of metalloproteinase 3 (TIMP3), a member of the TIMP family, participates in various inflammatory diseases, including liver ischemia/reperfusion injury (19), osteoarthritis (20) and IPF (21). Notably, miR-222-3p may directly target TIMP3 in osteosarcoma (22). However, the potential regulatory mechanism of GAS5 associated with the miR-222-3p/TIMP3 axis in MPP remains unknown.

The present study investigated the expression of GAS5, miR-222-3p and TIMP3 in patients with MPP. Lipid-associated membrane proteins (LAMPs) were induced in THP-1 cells to model MPP. The associations among GAS5, miR-222-3p and TIMP3 were confirmed. Subsequently, whether GAS5 controlled the viability and inflammation of LAMP-induced THP-1 cells by regulating the miR-222-3p/TIMP3 axis was investigated. The findings indicated a potential novel therapeutic target for MPP.

Materials and methods

Patients and samples

A total of 25 children with MPP from the Pediatric Intensive Care Unit of the Liaocheng Second People's Hospital (Linqing, China) were enrolled between July 2017 and October 2018. Exclusion criteria included premature delivery, immunodeficiency, recurrent pneumonia and recent use of immunomodulators and immunosuppressive agents. There were 16 children with PE and nine children without PE. Additionally, 25 healthy children comprised the control group. The healthy children had no history of MPP, immune system diseases, or other acute and chronic infectious diseases. Peripheral blood samples were collected from children in the MPP and control groups. A portion of the peripheral blood samples were used for the measurement of inflammatory cytokines and another portion was used to extract peripheral blood mononuclear cells (PBMCs). Next, the PBMCs were isolated by Ficoll-Plaque density gradient centrifugation (GE Healthcare) from all blood samples at 1,000 × g for 30 min at 20°C. The PBMC layer was extracted and washed by adding three volumes of PBS, centrifuged at 250 × g for 10 min at 20°C. The PBMCs were maintained in RPMI-1640 medium (Invitrogen; Thermo Fisher Scientific, Inc.), containing 10% fetal bovine serum (FBS; Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C in an atmosphere of 5% CO2. Logarithmic growth phase cells were used for further assays. The present study was conducted in accordance with the Declaration of Helsinki and was approved by the Ethics Committee of the Liaocheng Second People's Hospital (approval no. 2017–013). Written informed consent was obtained from each child and their guardian.

Extraction of LAMPs from MP

MP M129 [29342; American Type Culture Collection (ATCC)] was used as the standard MP strain. The MP was cultured in PPLO broth medium (BioLife) at 37°C for 5–7 days and harvested when the red pH indicator turned orange. Following centrifugation at 10,000 × g for 20 min at 4°C, MP pellets were resuspended in 10 ml Tris-buffered saline (TBS; 50 mM Tris pH 8.0, 0.15 M NaCl), containing 1 mM EDTA (TBSE). MP was then lysed for 1 h at 4°C by adding 2% (v/v) Triton X-114 into the suspension. To isolate LAMPs, the lysate was incubated for 10 min at 37°C to allow phase separation. The upper aqueous phase was removed and replaced with the same volume of TBSE. The phase separation procedure was repeated twice. The final phase was resuspended in TBSE to the original volume, and 2.5 volumes of ethanol were added to precipitate LAMPs at −20°C overnight. After the supernatant was discarded, the isolated LAMPs were resuspended in PBS, treated with low temperature ultrasound and stored at −70°C. The concentration of LAMPs was determined using an enhanced BCA Protein assay kit (Beyotime Institute of Biotechnology).

Cell culture and treatment

THP-1 cells (1×105 cells/ml), obtained from ATCC, were cultured in RPMI-1640 medium containing 10% FBS (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C in an atmosphere of 5% CO2. THP-1 cells were harvested and centrifuged after reaching exponential growth. The resulting cell pellet was suspended in serum-free RPMI-1640 medium, 1×106 cells were distributed across a 24-well plate. The cells were co-cultured with different levels of LAMPs (2, 4 and 6 µg/ml) for 16 h. Cells in the control group were treated with PBS. THP-1 cells co-cultured with 6 µg/ml LAMPs were considered as LAMP-induced THP-1 cells and were examined in subsequent experiments.

Cell transfection

Short hairpin (sh)-GAS5, sh-negative control (NC) and sh-TIMP3 were synthesized by Shanghai GenePharma Co., Ltd., and then inserted into the pGLVU6/Puro vector (Shanghai GenePharma Co., Ltd.). The pcDNA3.1-NC (pcDNA3.1), pcDNA3.1-GAS5 (oe-GAS5), miR-222-3p mimics (sense, 5′-AGCUACAUCUGGCUACUGGGU-3′ and antisense, 5′-CCAGUAGCCAGAUGUAGCUUU-3′), NC mimics (sense, 5′-UUCUCCGAACGUGUCACGUTT-3′ and antisense, 5′-ACGUGACACGUUCGGAGAATT-3′), miR-222-3p inhibitor (5′-ACCCAGUAGCCAGAUGUAGCU-3′), and NC inhibitor (5′-CAGUACUUUUGUGUAGUACAA-3′) were synthesized by Shanghai GenePharma Co., Ltd. LAMP-induced THP-1 cells (1×105 cells/well) were seeded onto a 24-well plate and cultured until growth was 80% confluent. The lipid complex was pre-prepared by mixing 25 µl Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) diluted in serum-free RPMI-1640 medium and 25 µl specific nucleic acids (sh-GAS5/sh-TIMP3/sh-NC/oe-GAS/pcDNA3.1 and/or miR-222-3p mimics or mimics NC/miR-222-3p inhibitor/inhibitor NC; shRNA, 200 ng; miRNA, 50 nM) diluted in serum-free medium for 10 min. Cells were then incubated with 50 µl lipid complex for 48 h at 37°C. After 48 h transfection, the transfected cells were used for subsequent assays.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The cDNA samples were obtained through reverse transcription (37°C for 15 min; 85°C for 5 sec) using the PrimeScript RT Reagent kit (Takara Bio, Inc.). miScript SYBR Green PCR kit (Qiagen, Inc.) was used to conduct a qPCR analysis. RT-qPCR was performed using a 7500 Real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) under the following conditions: 95°C for 3 min and 40 cycles of 95°C for 15 sec and 60°C for 30 sec, and a final extension step at 72°C for 10 min. Relative expression was calculated using the 2−ΔΔCq method (23). GAPDH, U6 and β-actin were used for the normalization of GAS5, miR-222-3p and TIMP3, respectively. The primer sequences are presented in Table I.

Table I.

Primer sequences.

Table I.

Primer sequences.

Name of primerSequence (5′-3′)
GAS5-F CTTCTGGGCTCAAGTGATCCT
GAS5-R TTGTGCCATGAGACTCCATCAG
GAPDH-F CGACTTATACATGGCCTTA
GAPDH-R TTCCGATCACTGTTGGAAT
miR-222-3p-F AGCTACATCTGGCTACTGGGT
miR-222-3p-R GCGAGCACAGAATTAATACGAC
U6-F CTCGCTTCGGCAGCACA
U6-R AACGCTTCACGAATTTGCGT
TIMP3-F ACCGAGGCTTCACCAAGATG
TIMP3-R CATCATAGACGCGACCTGTCA
β-actin-F TGGAATCCTGTGGCATCCATGAAAC
β-actin-R ACGCAGCTCAGTAACAGTCCG

[i] F, forward; R, reverse; GAS5, growth arrest-specific 5; TIMP3, tissue inhibitor of metalloproteinases-3.

Western blot analysis

A Nuclear and Cytoplasmic Protein Extraction kit (Beyotime Institute of Biotechnology) was used to extract nuclear and cytoplasmic proteins. PBMCs were lysed using RIPA lysis buffer (Beyotime Institute of Biotechnology) on ice to extract the total protein. Protein concentration was evaluated by bicinchoninic acid assay (Beyotime Institute of Biotechnology). Equal amounts (30 µg) of protein samples were separated by 10% SDS-PAGE. The separated proteins were transferred to polyvinylidene fluoride membranes, blocked with 5% skimmed milk for 1 h at 37°C, and incubated at 4°C overnight with primary antibodies against anti-TIMP3 (1:1,000; cat. no. SAB4502973; Sigma-Aldrich; Merck KGaA) or anti-Tubulin (1:200; cat. no. T3526; Sigma-Aldrich; Merck KGaA). The membranes were then incubated with a horseradish peroxidase-labeled goat anti-rabbit IgG (1:5,000; cat. no. 12-348; Sigma-Aldrich; Merck KGaA) secondary antibody for 1 h at 25°C. Finally, the bands were visualized using an enhanced chemiluminescence kit (Invitrogen; Thermo Fisher Scientific, Inc.). Protein bands were semi-quantified by densitometric analysis using ImageJ software (version 1.51, National Institutes of Health).

Viability assay

The viability of LAMP-induced THP-1 cells was measured using an MTT cell proliferation assay kit (Sigma-Aldrich; Merck KGaA). In brief, the LAMP-induced THP-1 cells with different transfections were seeded onto 96-well plates (2×103 cells/well) and cultured with 5% CO2 at 37°C. When the cell reached 80% confluence, they were incubated with 20 µg MTT reagents for 2 h at 37°C in a humidified culture chamber supplied with 5% CO2. Following incubation, supernatants were removed and 150 µl DMSO was added to dissolve the formazan crystals. The optical density values were measured at 450 nm using a plate reader and were used to evaluate cell viability.

ELISA

Peripheral blood samples from each group were transferred to a serum separating tube and centrifuged at 1,000 × g at 4°C for 10 min. The serum was harvested. Additionally, LAMP-induced THP-1 cells from each group were centrifuged at 1,000 × g at 4°C for 10 min. Each supernatant was collected. The levels of interleukin (IL)-6 and interferon (IFN)-γ in serum were measured using a Human IL-6 ELISA kit (cat. no. ab178013; Abcam) and Human IFN-γ ELISA kit (cat. no. ab46025; Abcam). The levels of IL-1β, IL-6, tumor necrosis factor-α (TNF-α) and heme oxygenase-1 (HO-1) in LAMP-induced THP-1 cells were assessed using Human IL-1β ELISA kit (cat. no. ab214025; Abcam), Human IL-6 ELISA kit (cat. no. ab178013; Abcam), Human TNF-α ELISA kit (cat. no. ab181421; Abcam) and Human HO-1 ELISA kit (cat. no. ab207621; Abcam), respectively.

Dual-luciferase reporter assay

The potential binding sites of GAS5 and miR-222-3p or miR-222-3p and TIMP3 were predicted by starBase (http://starbase.sysu.edu.cn/starbase2/) or TargetScan (http://www.targetscan.org/vert_72/). Wild-type (WT) fragments of the 3′-UTR of GAS5/TIMP3 with putative binding sites of miR-222-3p were purchased from Shanghai GenePharma Co., Ltd. and cloned into a psiCHECK-2 Dual-Luciferase miRNA Target Expression Vector (Promega Corporation). GAS5/TIMP3-3′-UTR-Mut reporter containing mutant miR-222-3p binding sites was used and generated using a Quikchange Multi Site-directed Mutagenesis kit (Stratagene; Agilent Technologies, Inc.). Subsequently, the recombinant vectors were co-transfected with miR-NC or miR-222-3p mimics into THP-1 cells. Luciferase activity was evaluated 48 h post-transfection by Dual Luciferase Reporter assay system (Promega Corporation), and firefly luciferase activity was normalized to that of Renilla luciferase.

Statistical analyses

All statistical analyses were performed using GraphPad Prism 7.0 (GraphPad Software, Inc.). Data are presented as the mean ± standard deviation. The differences between two groups or among multiple groups were assessed using Student's t-test or one-way analysis of variance followed by Tukey's post-hoc test. The significance of the correlations was determined by Pearson's correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Decreased GAS5 expression in PBMCs of patients with MPP

The demographic and clinical characteristics of the children with MPP and control groups are presented in Table II. Age, sex and white blood cell counts were not significantly different between the MPP and control groups. The levels of neutrophils and C-reactive protein in children with MPP were significantly higher than those of the control group (P<0.001). RT-qPCR was performed to confirm whether GAS5 was differentially expressed in the PBMCs of patients with MPP. GAS5 expression was significantly decreased in PBMCs of the MPP group compared with the control group (P<0.001; Fig. 1A). Additionally, GAS5 expression was significantly decreased in MPP cases with PE compared with those without PE (P<0.001; Fig. 1B). The level of GAS5 expressed by THP-1 cells significantly decreased in a dose-dependent manner following induction with LAMPs (P<0.001; Fig. 1C). Ten peripheral blood samples were randomly selected from control and MMP groups, respectively. Next, the levels of IL-6 and IFN-γ in serum were measured by ELISA. The results demonstrated that the levels of IL-6 and IFN-γ in serum in the MMP group were higher than those in the control group (P<0.001; Fig. 1D).

Table II.

Demographic and clinical characteristics of children with MPP and controls.

Table II.

Demographic and clinical characteristics of children with MPP and controls.

ParameterMPP (n=25)Control (n=25)P-value
Age, mean ± SD, years6.5±2.86.7±2.8   0.786
Sex, male/female, n13/1214/11   0.365
White blood cells, mean ± SD, ×109/l8.2±3.77.6±1.9   0.126
Neutrophils mean ± SD, ×109/l7.9±2.61.9±0.7<0.001
C-reactive protein, 25th-75th percentile, mg/l17.1 (9.8–43.5)0.16 (0.09–0.7)<0.001
Lactate dehydrogenase, mean ± SD, U/l456.4±150.5
Lymphocytes subgroups, mean ± SD, %
CD3+67.9±8.6
CD3+CD4+36.7±7.1
CD3+CD8+27.6±6.9
CD3CD19+18.5±5.1
CD3CD (16+ 56+)12.8±6.5
CD19+CD23+9.1±4.4

[i] MPP, Mycoplasma pneumoniae pneumonia; SD, standard deviation.

GAS5 enhances the viability and inhibits the inflammation of LAMP-induced THP-1 cells

To investigate the effect of GAS5 on MPP in vitro, LAMP-induced THP-1 cells were used as the MPP model at the cellular level. As demonstrated in Fig. 2A, GAS5 expression was significantly inhibited or enhanced by the transfection of sh-GAS5 or oe-GAS5 into LAMP-induced THP-1 cells (P<0.001). The MTT assay revealed that GAS5-silencing significantly decreased the viability (P<0.01), while GAS5-overexpression significantly increased the viability of LAMP-induced THP-1 cells (P<0.001; Fig. 2B). The levels of IL-1β, IL-6 and TNF-α in LAMP-induced THP-1 cells were significantly increased by GAS5 silencing and decreased when GAS5 was overexpressed (P<0.001; Fig. 2C-E). Transfection of sh-GAS5 or oe-GAS5 significantly downregulated or upregulated the level of the anti-inflammatory cytokine HO-1 in LAMP-induced THP-1 cells (P<0.001; Fig. 2F).

miR-222-3p as a target of GAS5

Using the starBase online software, it was found that GAS5 contained the target site of miR-222-3p (Fig. 3A). A dual-luciferase reporter assay confirmed that miR-222-3p mimics reintroduction clearly decreased the luciferase activity in THP-1 cells transfected with GAS5 WT compared with the activity observed in miR-NC (P<0.001; Fig. 3B). RT-qPCR revealed that miR-222-3p expression was significantly enhanced in PBMCs of patients with MPP (P<0.001; Fig. 3C). As demonstrated in Fig. 3D, a negative correlation between GAS5 and miR-222-3p expression was observed in PBMCs of all participants (R2=0.556, P<0.0001). miR-222-3p was highly expressed in patients with MPP with PE (P<0.001; Fig. 3E). Notably, downregulation or upregulation of GAS5 significantly increased or decreased, respectively, miR-222-3p expression in LAMP-induced THP-1 cells (P<0.001; Fig. 3F).

miR-222-3p decreases the viability and increases the inflammation of LAMP-induced THP-1 cells

As demonstrated in Fig. 4A, miR-222-3p was enhanced or inhibited by the transfection of miR-222-3p mimics or miR-222-3p inhibitors, respectively, into LAMP-induced THP-1 cells (P<0.001). The overexpression of miR-222-3p significantly decreased viability (P<0.01), while its inhibition significantly increased the viability of LAMP-induced THP-1 cells (P<0.001; Fig. 4B). miR-222-3p-overexpression or inhibition increased or decreased, respectively, the levels of IL-1β, IL-6 and TNF-α in LAMP-induced THP-1 cells (P<0.001; Fig. 4C-E). Furthermore, the level of HO-1 in LAMP-induced THP-1 cells was significantly decreased or increased by the upregulation or downregulation, respectively, of miR-222-3p (P<0.001; Fig. 4F).

Targeting of TIMP3 by miR-222-3p

TargetScan was used to predict the binding site for miR-222-3p on the 3′-UTR of TIMP3 (Fig. 5A). miR-222-3p mimics notably decreased the luciferase activity of the WT TIMP3 reporter vector in THP-1 cells (P<0.001; Fig. 5B). Additionally, RT-qPCR demonstrated that TIMP3 expression was significantly downregulated in PBMCs of patients with MPP (P<0.001; Fig. 5C). As illustrated in Fig. 5D, the expression of TIMP3 was negatively correlated with miR-222-3p expression in PBMCs of all participants (R2=0.787, P<0.0001). Notably, TIMP3 expression was significantly suppressed in patients with MPP with PE (P<0.001; Fig. 5E). Furthermore, western blot analysis revealed that transfection of miR-222-3p mimics or miR-222-3p inhibitor may suppress or promote, respectively, the protein expression of TIMP3 in LAMP-induced THP-1 cells (P<0.001; Fig. 5F).

GAS5 increases the viability and attenuates the inflammation of LAMP-induced THP-1 cells by regulating the miR-222-3p/TIMP3 axis

As demonstrated in Fig. 6A, the expression of TIMP3 was significantly inhibited by transfecting with sh-TIMP3 in LAMP-induced THP-1 cells (P<0.001). To investigate whether GAS5 regulated the miR-222-3p/TIMP3 axis in MPP, feedback experiments were performed in LAMP-induced THP-1 cells. GAS5-overexpression notably increased TIMP3 protein expression, and miR-222-3p-overexpression or TIMP3-knockdown rescued the increased TIMP3 protein expression caused by GAS5-overexpression in LAMP-induced THP-1 cells (P<0.001; Fig. 6B). GAS5-overexpression increased cell viability (P<0.001). The upregulation of miR-222-3p or downregulation of TIMP3 reversed the promotion effect of GAS5-overexpression on the viability of LAMP-induced THP-1 cells (P<0.05; Fig. 6C). GAS5-overexpression significantly decreased the levels of IL-1β, IL-6 and TNF-α, and increased the level of HO-1 in LAMP-induced THP-1 cells (P<0.001; Fig. 6D-G). Notably, miR-222-3p-overexpression or TIMP-knockdown mitigated the inhibitory effect of GAS5-overexpression on the levels of the pro-inflammatory cytokines and weakened the promotion effect of GAS5-overexpression on the level of anti-inflammatory cytokines in LAMP-induced THP-1 cells (P<0.001).

Discussion

Downregulation of lncRNAs is associated with the pathogenesis of respiratory diseases. GAS5 expression is downregulated in diverse respiratory diseases, including acute respiratory distress syndrome (24), non-small cell lung cancer (25) and ALI (12). In the present study, GAS5 expression was markedly decreased in PBMCs of patients with MPP and in LAMP-induced THP-1 cells. Furthermore, the results of the present study demonstrated that GAS5 may be associated with the development of MPP. LncRNAs serve as critical factors to modulate cell viability in certain inflammatory diseases. LncRNA CASC9 may mitigate the effect of LPS on the viability of human small airway epithelial cells (26). LncRNA TUG1 suppressed MRC-5 cell viability in a pneumonia model (27). Notably, GAS5 increases the viability of endothelial progenitor cells, attenuating the development of atherosclerosis (28). In the present study, GAS5-overexpression increased the viability of LAMP-induced THP-1 cells, suggesting that GAS5 may increase cell viability in MPP. Certain lncRNAs are involved in the regulation of inflammation in lung diseases. In sepsis-induced ALI, lncRNA TUG1 ameliorates inflammation by inhibiting miR-34b-5p (29). MALAT1-silencing increases IL-6 and TNF-α levels in ALI rats (30). Notably, GAS5 has an anti-inflammatory role in numerous diseases. GAS5 regulates miR-429 to increase DUSP1 expression, attenuating the inflammation of alveolar epithelial cells in ALI (12). GAS5 inhibits endoplasmic reticulum stress-induced inflammation by targeting SERCA2b in diabetic retinopathy (31). In the present study, GAS5-overexpression suppressed the levels of IL-1β, IL-6 and TNF-α, and enhanced the HO-1 level in LAMP-induced THP-1 cells. These results indicated that GAS5 may attenuate inflammation in MPP. By contrast, GAS5 has a pro-inflammatory role in certain diseases. For instance, GAS5 accelerates microglial inflammatory responses in Parkinson's disease (32). Silencing of GAS5 decreases the levels of TNF-α and IL-6 in the supernatant of osteoarthritic chondrocytes (33). GAS5 may exert different regulatory effects on inflammation in different diseases. The regulatory role of GAS5 in inflammation is controversial and further research on the detailed regulatory mechanism of GAS5 on the inflammation of MPP is required.

Previous studies have demonstrated that GAS5 may interact with miRNAs to modulate the development of inflammatory diseases (34,35). For instance, GAS5 attenuates inflammation in systemic lupus erythematosus by suppressing miR-92a-3p expression (36). GAS5 decreases TNF-α and IL-6 levels in diabetic nephropathy by inhibiting miR-452-5p expression (37). In the present study, miR-222-3p was determined to be a target of GAS5. miR-222-3p expression was upregulated in patients with MMP compared with the children in the control group. Notably, miR-222-3p expression was negatively correlated with GAS5 expression in patients with MPP. We hypothesized that GAS5 may influence MPP by regulating miR-222-3p. miR-222 is upregulated and promotes the pathogenesis of inflammatory diseases. For instance, miR-222 accelerates endothelial cell inflammation and dysfunction in atherosclerosis (38). miR-222 is highly expressed and induces the inflammatory responses of microglia cells in intracerebral hemorrhage (39). Notably, miR-222-silencing attenuates staphylococcal enterotoxin B-induced inflammation in ALI (40). In the present study, miR-222-3p decreased the viability and accelerated the inflammation of LAMP-induced THP-1 cells. Furthermore, miR-222-3p markedly reversed the effects of GAS5-overexpression on LAMP-induced THP-1 cells. These results suggested that GAS5 may alleviate MPP by inhibiting miR-222-3p expression in vitro.

TIMP3 expression is usually downregulated in diverse inflammatory diseases, including heart failure (41), interstitial lung disease (42) and cardiac ischemia/reperfusion injury (43). Similarly, TIMP3 expression was clearly decreased in MPP and LAMP-induced THP-1 cells in the present study. TIMP3 generally exerts its role in certain diseases by interacting with miRNAs. For instance, miR-712 facilitates endothelial inflammation and atherosclerosis by inhibiting TIMP3 expression (44). miR-34b-5p-silencing hampers bleomycin-induced pulmonary fibrosis by increasing TIMP3 expression (21). Notably, miR-222 inhibition enhances TIMP3 expression to alleviate the development of pulmonary artery hypertension (45). In the present study, TIMP3 was a target of miR-222-3p. The expression of TIMP3 was lower in patients with MMP than in the children in control group. Notably, there was a negative correlation between the expression of TIMP3 and miR-222-3p in patients with MPP. We hypothesized that miR-222-3p may be involved in MPP development by modulating TIMP3. Considering the interaction between GAS5 and miR-222-3p, we hypothesized that silencing of GAS5 may downregulate TIMP3 expression by increasing miR-222-3p expression in MMP. The feedback data demonstrated that TIMP3-knockdown attenuated the enhancing effect on cell viability and weakened the anti-inflammatory effect caused by GAS5-overexpression in LAMP-induced THP-1 cells. These findings suggested that GAS5 may exert its protective effect by regulating the miR-222-3p/TIMP3 axis in MPP.

There were two limitations to the present study. To begin with, the detailed regulatory mechanism of GAS5 on the inflammation of MPP is not fully understood. Furthermore, the effect of GAS5 on MPP was investigated at the cellular level. Establishment of an MPP animal model is required to confirm the role of GAS5 in vivo.

In conclusion, the expression of GAS5 was downregulated in patients with MPP and LAMP-induced THP-1 cells. GAS5-overexpression increased the viability and attenuated the inflammation of LAMP-induced THP-1 cells by regulating the miR-222-3p/TIMP3 axis. These results indicated that GAS5 may be a promising therapeutic target for MPP.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

LY and XL were responsible for conceptualization and design of the study, and performed the methods and software analysis, as well as having roles in the supervision of the entire study. XZ was responsible for data acquisition, original draft preparation and project management. XZ and XL were responsible for reviewing and editing the manuscript. LY, XZ and XL confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by The Ethics Committee of The Second People's Hospital of Liaocheng (Linqing, China) and written informed consent was obtained from each child and their guardian.

Patient consent for publication

Written informed consent for publication was obtained from each child and their guardian.

Competing interests

The authors declare that they have no competing interests.

References

1 

Saraya T: The history of Mycoplasma pneumoniae pneumonia. Front Microbiol. 7:3642016. View Article : Google Scholar : PubMed/NCBI

2 

Søndergaard M, Friis MB, Hansen DS and Jørgensen IM: Clinical manifestations in infants and children with Mycoplasma pneumoniae infection. PLoS One. 13:e01952882018. View Article : Google Scholar : PubMed/NCBI

3 

Izumikawa K, Izumikawa K, Takazono T, Kosai K, Morinaga Y, Nakamura S, Kurihara S, Imamura Y, Miyazaki T, Tsukamoto M, et al: Clinical features, risk factors and treatment of fulminant Mycoplasma pneumoniae pneumonia: A review of the Japanese literature. J Infect Chemother. 20:181–185. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Izumikawa K: Clinical features of severe or fatal Mycoplasma pneumoniae pneumonia. Front Microbiol. 7:8002016. View Article : Google Scholar : PubMed/NCBI

5 

Lee H, Yun KW, Lee HJ and Choi EH: Antimicrobial therapy of macrolide-resistant Mycoplasma pneumoniae pneumonia in children. Expert Rev Anti Infect Ther. 16:23–34. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Lee E, Cho HJ, Hong SJ, Lee J, Sung H and Yu J: Prevalence and clinical manifestations of macrolide resistant Mycoplasma pneumoniae pneumonia in Korean children. Korean J Pediatr. 60:151–157. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Du Y, Hao X and Liu X: Low expression of long noncoding RNA CDKN2B-AS1 in patients with idiopathic pulmonary fibrosis predicts lung cancer by regulating the p53-signaling pathway. Oncol Lett. 15:4912–4918. 2018.PubMed/NCBI

8 

Dai L, Zhang G, Cheng Z, Wang X, Jia L, Jing X, Wang H, Zhang R, Liu M, Jiang T, et al: Knockdown of LncRNA MALAT1 contributes to the suppression of inflammatory responses by up-regulating miR-146a in LPS-induced acute lung injury. Connect Tissue Res. 59:581–592. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Huang S, Feng C, Chen L, Huang Z, Zhou X, Li B, Wang LL, Chen W, Lv FQ and Li TS: Identification of potential key long non-coding RNAs and target genes associated with pneumonia using long non-coding RNA sequencing (lncRNA-Seq): A preliminary study. Med Sci Monit. 22:3394–3408. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Zhang L, Zhao S and Zhu Y: Long noncoding RNA growth arrest-specific transcript 5 alleviates renal fibrosis in diabetic nephropathy by downregulating matrix metalloproteinase 9 through recruitment of enhancer of zeste homolog 2. FASEB J. 34:2703–2714. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Li F, Sun J, Huang S, Su G and Pi G: LncRNA GAS5 overexpression reverses LPS-induced inflammatory injury and apoptosis through up-regulating KLF2 expression in ATDC5 chondrocytes. Cell Physiol Biochem. 45:1241–1251. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Li J and Liu S: LncRNA GAS5 suppresses inflammatory responses and apoptosis of alveolar epithelial cells by targeting miR-429/DUSP1. Exp Mol Pathol. 113:1043572020. View Article : Google Scholar : PubMed/NCBI

13 

Pan J, Ye Z, Zhang N, Lou T and Cao Z: MicroRNA-217 regulates interstitial pneumonia via IL-6. Biotechnol Biotechnol Equip. 32:1541–1547. 2018. View Article : Google Scholar

14 

Wang Z, Zheng Y, Fang Z and Zhang Y: The role of miR-21 and its predicted target gene, PTEN, in the development of ventilator associated pneumonia. Biomed Res. 28:3967–3973. 2017.

15 

Podsiad A, Standiford TJ, Ballinger MN, Eakin R, Park P, Kunkel SL, Moore BB and Bhan U: MicroRNA-155 regulates host immune response to postviral bacterial pneumonia via IL-23/IL-17 pathway. Am J Physiol Lung Cell Mol Physiol. 310:L465–L475. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Chu C, Lei X, Li Y, Luo Y, Ding Y, Zhou W and Ji W: High expression of miR-222-3p in children with Mycoplasma pneumoniae pneumonia. Ital J Pediatr. 45:1632019. View Article : Google Scholar : PubMed/NCBI

17 

Zhang X, Ye Y and Zhao S: LncRNA Gas5 acts as a ceRNA to regulate PTEN expression by sponging miR-222-3p in papillary thyroid carcinoma. Oncotarget. 9:3519–3530. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Muniategui A, Nogalescadenas R, Vazquez M, Aranguren XL, Agirre X, Luttun A, Prosper F, Pascual-Montano A and Rubio A: Quantification of miRNA-mRNA interactions. PLoS One. 7:e307662012. View Article : Google Scholar : PubMed/NCBI

19 

Fujii T, Duarte S, Lee E, Ke B, Busuttil RW and Coito AJ: Tissue inhibitor of metalloproteinase 3 deficiency disrupts the hepatocyte E-cadherin/β-catenin complex and induces cell death in liver ischemia/reperfusion injury. Liver Transpl. 26:113–126. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Nakamura H, Vo P, Kanakis I, Liu K and Bou-Gharios G: Aggrecanase-selective tissue inhibitor of metalloproteinase-3 (TIMP3) protects articular cartilage in a surgical mouse model of osteoarthritis. Sci Rep. 10:92882020. View Article : Google Scholar : PubMed/NCBI

21 

Hu RP, Lu YY and Zhang XJ: MiR-34b-5p knockdown attenuates bleomycin-induced pulmonary fibrosis by targeting tissue inhibitor of metalloproteinase 3 (TIMP3). Eur Rev Med Pharmacol Sci. 23:2273–2279. 2019.PubMed/NCBI

22 

Guo J, Liu Q, Li Z, Guo H, Bai C and Wang F: miR-222-3p promotes osteosarcoma cell migration and invasion through targeting TIMP3. Onco Targets Ther. 11:8643–8653. 2018. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Li HB, Zi PP, Shi HJ, Gao M and Sun RQ: Role of signaling pathway of long non-coding RNA growth arrest-specific transcript 5/microRNA-200c-3p/angiotensin converting enzyme 2 in the apoptosis of human lung epithelial cell A549 in acute respiratory distress syndrome. Zhonghua Yi Xue Za Zhi. 98:3354–3359. 2018.(In Chinese). PubMed/NCBI

25 

Wu Y, Lyu H, Liu H, Shi X, Song Y and Liu B: Downregulation of the long noncoding RNA GAS5-AS1 contributes to tumor metastasis in non-small cell lung cancer. Sci Rep. 6:310932016. View Article : Google Scholar : PubMed/NCBI

26 

Wang HR, Guo XY, Liu XY and Song X: Down-regulation of lncRNA CASC9 aggravates sepsis-induced acute lung injury by regulating miR-195-5p/PDK4 axis. Inflamm Res. 69:559–568. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Meng J, Chen Y and Zhang C: Protective impacts of long noncoding RNA taurine-upregulated 1 against lipopolysaccharide-evoked injury in MRC-5 cells through inhibition of microRNA-127. J Cell Biochem. 120:14928–14935. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Yao J, Shi Z, Ma X, Xu D and Ming G: lncRNA GAS5/miR-223/NAMPT axis modulates the cell proliferation and senescence of endothelial progenitor cells through PI3K/AKT signaling. J Cell Biochem. 120:14518–14530. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Qiu N, Xu X and He Y: LncRNA TUG1 alleviates sepsis-induced acute lung injury by targeting miR-34b-5p/GAB1. BMC Pulm Med. 20:492020. View Article : Google Scholar : PubMed/NCBI

30 

Li H, Shi H, Ma N, Zi P, Liu Q and Sun R: BML-111 alleviates acute lung injury through regulating the expression of lncRNA MALAT1. Arch Biochem Biophys. 649:15–21. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Jiang L, Wang C and Shen X: LncRNA GAS5 suppresses ER stress-induced apoptosis and inflammation by regulating SERCA2b in HG-treated retinal epithelial cell. Mol Med Rep. 22:1072–1080. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Xu W, Zhang L, Geng Y, Liu Y and Zhang N: Long noncoding RNA GAS5 promotes microglial inflammatory response in Parkinson's disease by regulating NLRP3 pathway through sponging miR-223-3p. Int Immunopharmacol. 85:1066142020. View Article : Google Scholar : PubMed/NCBI

33 

Ji Q, Qiao X, Liu Y, Wang D and Yan J: Silencing of long-chain non-coding RNA GAS5 in osteoarthritic chondrocytes is mediated by targeting the miR-34a/Bcl-2 axis. Mol Med Rep. 21:1310–1319. 2020.PubMed/NCBI

34 

Li G, Du P, Qiang X, Jin D, Liu H, Li B and Guo J: Low-expressed GAS5 injure myocardial cells and progression of chronic heart failure via regulation of miR-223-3P. Exp Mol Pathol. 117:1045292020. View Article : Google Scholar : PubMed/NCBI

35 

Shangguan Y, Han J and Su H: GAS5 knockdown ameliorates apoptosis and inflammatory response by modulating miR-26b-5p/Smad1 axis in cerebral ischaemia/reperfusion injury. Behav Brain Res. 379:1123702020. View Article : Google Scholar : PubMed/NCBI

36 

Liu Q, Deng Y, Li C, Xie H, Liu Q, Ming S, Wu D and Luo F: LncRNA GAS5 suppresses CD4+ T cell activation by upregulating E4BP4 via inhibiting miR-92a-3p in systemic lupus erythematosus. Immunol Lett. 227:41–47. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Xie C, Wu W, Tang A, Luo N and Tan Y: lncRNA GAS5/miR-452-5p reduces oxidative stress and pyroptosis of high-glucose-stimulated renal tubular cells. Diabetes Metab Syndr Obes. 12:2609–2617. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Xue Y, Wei Z, Ding H, Wang Q, Zhou Z, Zheng S, Zhang Y, Hou D, Liu Y, Zen K, et al: MicroRNA-19b/221/222 induces endothelial cell dysfunction via suppression of PGC-1α in the progression of atherosclerosis. Atherosclerosis. 241:671–681. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Bai YY and Niu JZ: miR-222 regulates brain injury and inflammation following intracerebral hemorrhage by targeting ITGB8. Mol Med Rep. 21:1145–1153. 2020.PubMed/NCBI

40 

Chen L, Chen J, Xie G and Zhu L: MiR-222 inhibition alleviates Staphylococcal Enterotoxin B-induced inflammatory acute lung injury by targeting Foxo3. J Biosci. 45:652020. View Article : Google Scholar : PubMed/NCBI

41 

Kassiri Z, Oudit GY, Sanchez O, Dawood F, Mohammed FF, Nuttall RK, Edwards DR, Liu PP, Backx PH and Khokha R: Combination of tumor necrosis factor-alpha ablation and matrix metalloproteinase inhibition prevents heart failure after pressure overload in tissue inhibitor of metalloproteinase-3 knock-out mice. Circ Res. 97:380–390. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Nagar JK, Patel PP, Mohapatra JN, Sharma MM, Pandya GM, Umar MM, Chatterjee AA, Deshpande SS, Jain MR and Soni HM: Differential effects of dexamethasone and rosiglitazone in a sephadex-induced model of lung inflammation in rats: Possible role of tissue inhibitor of metalloproteinase-3. Indian J Pharmacol. 47:153–159. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Liu H, Jing X, Dong A, Bai B and Wang H: Overexpression of TIMP3 protects against cardiac ischemia/reperfusion injury by inhibiting myocardial apoptosis through ROS/Mapks pathway. Cell Physiol Biochem. 44:1011–1023. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Son DJ, Kumar S, Takabe W, Kim CW, Ni CW, Alberts-Grill N, Jang IH, Kim S, Kim W, Won Kang S, et al: The atypical mechanosensitive microRNA-712 derived from pre-ribosomal RNA induces endothelial inflammation and atherosclerosis. Nat Commun. 4:30002013. View Article : Google Scholar : PubMed/NCBI

45 

Xu Y, Bei Y, Shen S, Zhang J, Lu Y, Xiao J and Li X: MicroRNA-222 promotes the proliferation of pulmonary arterial smooth muscle cells by targeting P27 and TIMP3. Cell Physiol Biochem. 43:282–292. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 23 Issue 5

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
Yang L, Zhang X and Liu X: Long non‑coding RNA GAS5 protects against <em>Mycoplasma pneumoniae</em> pneumonia by regulating the microRNA‑222‑3p/TIMP3 axis. Mol Med Rep 23: 380, 2021
APA
Yang, L., Zhang, X., & Liu, X. (2021). Long non‑coding RNA GAS5 protects against <em>Mycoplasma pneumoniae</em> pneumonia by regulating the microRNA‑222‑3p/TIMP3 axis. Molecular Medicine Reports, 23, 380. https://doi.org/10.3892/mmr.2021.12019
MLA
Yang, L., Zhang, X., Liu, X."Long non‑coding RNA GAS5 protects against <em>Mycoplasma pneumoniae</em> pneumonia by regulating the microRNA‑222‑3p/TIMP3 axis". Molecular Medicine Reports 23.5 (2021): 380.
Chicago
Yang, L., Zhang, X., Liu, X."Long non‑coding RNA GAS5 protects against <em>Mycoplasma pneumoniae</em> pneumonia by regulating the microRNA‑222‑3p/TIMP3 axis". Molecular Medicine Reports 23, no. 5 (2021): 380. https://doi.org/10.3892/mmr.2021.12019