Open Access

Identification of TLR2 and TLR4‑induced microRNAs in human mesenchymal stem cells and their possible roles in regulating TLR signals

  • Authors:
    • Xingbing Wang
    • Yunxia Zhu
    • Bo Xu
    • Jing Wang
    • Xin Liu
  • View Affiliations

  • Published online on: April 27, 2016     https://doi.org/10.3892/mmr.2016.5197
  • Pages: 4969-4980
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Toll‑like receptors (TLRs) are expressed in human bone marrow‑derived mesenchymal stromal cells (BM‑MSCs), and the activation of TLRs is important in proliferation, differentiation, migration and hematopoiesis‑supporting functions of BM‑MSCs. However, the molecular mechanisms underlying these processes remain to be elucidated. MicroRNAs (miRNAs) are involved in various biological functions by mediating mRNA degradation or inhibiting translation of target genes. The present study aimed to identify whether TLRs regulate the expression of miRNAs in BM‑MSCs and elucidate the regulatory roles of miRNAs. Illumina high‑throughput sequencing was used to profile miRNAs expressed in BM‑MSCs stimulated with TLR2 agonist, PAM3CSK4 (PM) or TLR4 agonist, lipopolysaccharides (LPS). A marked expression change upon PM or LPS treatment was observed for 164 known miRNAs and six novel miRNAs that were identified. The expression of six novel miRNAs and 40 randomly selected known miRNAs was further validated by reverse transcription‑quantitative polymerase chain reaction. In addition, bioinformatic methods were used to predict the potential target genes of the abundant known miRNAs. The gene ontology analysis demonstrated that predicted targets were enriched in the regulation of signal transduction, cellular processes and macromolecule metabolic processes. Kyoto Encyclopedia of Genes and Genomes pathway analysis suggested that these potential targets were involved in numerous important pathways, predominantly including mitogen‑activated protein kinase, phosphatidylinositol‑4,5‑bisphosphate 3‑kinase‑Akt, neurotrophin and cancer‑associated signaling pathways. The present study aimed to identify the global expression change of miRNAs in BM‑MSCs stimulated with LPS and PM, providing the opportunity to further elucidate the roles of miRNAs in mediating TLR signals to regulate the functions of BM‑MSCs.

Introduction

MicroRNAs (miRNAs) are a class of small non-coding RNAs (length, ~22 nt) that regulate gene expression at the post-transcriptional levels. miRNAs are involved in the regulation of the majority of important biological events, including differentiation, growth, proliferation, survival, signal transduction and immune response (13). However, the roles of miRNAs in regulating the activation of human bone marrow-derived mesenchymal stromal cells (BM-MSCs) remain to be elucidated.

The BM-MSCs are multipotent cells that differentiate into osteoblasts, adipocytes, chondrocytes and other tissue cells (47). It has been demonstrated that toll-like receptors (TLRs) are expressed in MSCs to modulate their proliferation, cytokine secretion, differentiation, hematopoiesis-supporting functions and immunosuppressive capacity (810). Notably, it is well established that TLRs induce multiple miRNAs, which regulate TLR-signaling responses at multiple levels. For example, TLR2 and TLR4 are repressed by expression of miR-105, miR-146 and the let-7 miRNA family (1114). However, miR-155 and miR-146b directly target numerous TLR signaling proteins (15,16). Regulatory molecules, TLR-induced transcription factors and the inflammatory cytokines are also regulated by miRNAs, including miR-155 (17).

It has previously been verified that pre-stimulation with the TLR2 agonist, PAM3CSK4 (PM) or the TLR4 agonist, lipopolysaccharides (LPS) enables BM-MSCs to enhance CD34+ cell proliferation and differentiation towards the myeloid lineage (10). To further elucidate the roles of miRNAs in mediating TLR-induced BM-MSC activation, the present study aimed to determine the miRNA expression profiles in unstimulated BM-MSCs and PM/LPS-stimulated BM-MSCs, using high-throughput Illumine HiSeq 2000 technology. Bioinformatic methods were also used to predict the potential target genes of the abundant known miRNAs. Furthermore, data from the present study indicated that miRNAs are involved in various important functions in human BM-MSCs, including the TLR signaling pathway.

Materials and methods

Isolation and culture of BM-MSCs

BM-MSCs were isolated from fresh bone marrow by sterile puncture of a healthy donor. The present study was approved by the Anhui Medical University Ethics Committee, and informed written consent was obtained from the donor. The isolation, expansion and identification of BM-MSCs were performed as described previously (10). Briefly, following density gradient centrifugation over Ficoll-Hypaque (1.077 g/ml; Beijing Solarbio Science &Technology Co., Ltd., Beijing, China), BM-MSCs were cultured (1×106 cells/ml) in Dulbecco's High Glucose Modified Eagles Medium (GE Healthcare Life Sciences, Logan, UT, USA), supplemented with 10% fetal calf serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and containing 5% CO2 at 37°C. Non-adherent cells were removed after 24 h and the culture medium was changed twice a week. Adherent cells were cultured until 80–90% confluence was reached, after which trypsin (0.25%; Gibco) digestion was conducted on passage three cells. BM-MSCs were analyzed by flow cytometry (FACSCalibur™; BD Biosciences, Franklin Lakes, NJ, USA) following staining with monoclonal antibodies, as follows: Fluorescein isothiocyanate (FITC)-conjugated anti-cluster of differentiation (CD) 90 (1:10; cat. no. 555595); FITC-anti-CD14 (1:10; cat. no. 555397), phycoerythrin (PE)-anti-CD29 (cat. no. 555443), cyanine5-PE-anti-CD34 (cat. no. 555823), PE-anti-CD166 (cat. no. 559263), FITC-anti-CD44 (1:10; cat. no. 555478), PE-anti-CD31 (1:10; cat. no. 560983), FITC-anti-CD45 (1:10; cat. no. 555482), PE-anti-CD13 (1:10; cat. no. 560998) and FITC-anti-CD105 (1:10; cat. no. 561443; all: BD Biosciences). BM-MSCs were confirmed by negative staining of hematopoietic and endothelial lineage markers (CD14, CD34, CD31 and CD45) and positive staining of CD90, CD105, CD166, CD29, CD44 and CD13.

PM and LPS stimulation

LPS (100 ng/ml) and PM (100 ng/ml) were purchased from R&D Systems, Inc. (Minneapolis, Minnesota, USA). The BM-MSC feeder layers in 12-well plates were stimulated with (experimental group) or without (control) TLR2 agonist, PM, or TLR4 agonist, LPS, for 24 h. The supernatants were removed and the feeder layers were washed twice with fresh culture medium.

Total RNA extraction and small RNA sequencing library preparation

Total RNA was isolated from confluent MSCs with TRIzol (Promega Corporation, Madison, WI, USA). The purity and concentration of total RNA samples were determined with NanoDrop ND-1000 (Thermo Fisher Scientific, Inc., Wilmington, DE, USA). Total RNA of each sample was used to prepare the miRNA sequencing library using the TruSeq Small RNA Library Prep kit (RS-200-0012; Illumina, Inc., San Diego, CA, USA), according to the manufacturer's protocol. This included the following steps: i) 3′-Adapter ligation with truncated T4 RNA ligase 2 (New England BioLabs, Inc., Ipswich, MA, USA); ii) 5′-adapter ligation with T4 RNA ligase (Ambion; Thermo Fisher Scientific, Inc.); iii) cDNA synthesis with reverse transcription primers (Bioligo Biological Technology, Co., Ltd., Shanghai, China); iv) polymerase chain reaction (PCR) amplification; and v) extraction and purification of PCR amplified fragments [length,~138–158 bp; corresponding to small RNAs (length,~15–35 nt)] from the agarose gel. Subsequently, the completed libraries were quantified with an Agilent 2100 Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA).

Small RNA sequencing

The samples were diluted to a final concentration of 8 pM and cluster generation was performed on the Illumina cBot using TruSeq Rapid SR cluster kit (Illumina, Inc.), following the manufacturer's protocols. Sequencing was performed on an Illumina HiSeq 2000 using TruSeq Rapid SBS kits (Illumina, Inc.), according to the manufacturer's protocols. The DNA fragments in the libraries were denatured with 0.1 M NaOH to generate single-stranded DNA molecules, captured on flow cells (Illumina, Inc.), amplified in situ and finally sequenced for 36 cycles on Illumina HiSeq 2000.

Analysis of sequencing data and length distribution

Using high throughput sequencing with an Illumina Hiseq 2000, total clean reads from the control and trial libraries were obtained, and the length distribution of the clean reads was summarized. Image analysis and base calling were performed using Off-Line Basecaller software (v1.8.0; Illumina, San Diego, CA, USA). Subsequently, the 3′-adapter sequence was trimmed from the clean reads [that had passed the Solexa CHASTITY quality filter (Illumina)] and the reads of length <15 nt were discarded. Remaining reads (length, ≥15 nt) were aligned to the latest known human reference miRNA precursor set [Sanger miRBase 19 (http://www.mirbase.org/)] using Novoalign software (v2.07.11; http://www.novocraft.com/products/novoalign/). Reads (<2 counts) were discarded when calculating the miRNA expression. In order to characterize the isomiR variability, any sequence that matched the miRNA precursors in the mature miRNA region ±4 nt (with ≤1 mismatch) were accepted as mature miRNA isomiRs, which were grouped according to the 5-prime (5p) or 3-prime (3p) arm of the precursor hairpin.

Prediction of novel miRNAs

miRDeep2 (http://www.mdc-berlin.de/en/research/research_teams/systems_biology_of_gene_regulatory_elements/projects/miRDeep) was used to predict novel miRNAs. For novel miRNA prediction, all sequence data was pooled from the following 3′-adapter trimmed files: LPS, PM and contrimmed_tags.fa, all adapter trimmed sequences of length <17 bp and mismatch >1 were excluded from the prediction pipeline. The higher the novel miRNA score of the miRDeep2, the more reliable the novel miRNA was considered to be.

Target gene prediction

Three online search algorithms, TargetScan version 6.2 (http://www.targetscan.org/vert_60/) and miRanda (http://www.Microrna.org/microrna/home.do) and MicroCosm Targets version 5 (http://www.ebi.ac.uk/enright-srv/microcosm/htdocs/targets/v5/) were used to predict the target genes of differentially expressed (DE) miRNAs among the BM-MSCs activated with the TLR2 and TLR4 agonists, and BM-MSCs in the absence of agonists. The annotated miRNA target genes that were selected from all the algorithms were considered to be the target genes.

Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the predicted miRNA target genes

In order to further realize the functions, the predicted target genes were subjected to the analysis of GO project (http://www.geneontology.org). Fisher's exact test was used to find whether there was increased overlap between the DE list and the GO annotation list than would be expected by chance. The P-value denotes the significance of GO term enrichment in the DE genes; the lower the P-value, the more significant the GO term (P<0.05 is recommended). Furthermore, pathway analysis was performed for these target genes. Pathway analysis is a functional analysis that maps genes to the KEGG pathways. The P-value (EASE-score, Fisher's method P-value or hypergeometric P-value) indicates the significance of the pathway correlated to the conditions. A lower P-value, indicates a more significant pathway (P<0.05 is recommended).

Validation of miRNA expression by quantitative PCR (qPCR)

A random selection of DE miRNAs between the experimental and control groups from the sequencing data was validated by qPCR. Corroboration of the six novel miRNAs was performed according to the previously described conditions using qPCR assays. Total RNA was isolated from each sample using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The miRNA quantification was performed by qPCR using Applied Biosystems StepOne Real-Time PCR system (Thermo Fisher Scientific, Inc.) and SYBR premix Ex Taq II kit (Takara Bio, Inc.) according to the manufacturer's protocols. U6 served to normalize the levels of all miRNA transcripts, the primer sequences were forward, 5′-GCT TCG GCA GCA CAT ATA CTA AAAT-3′ and reverse, 5′-CGC TTC ACG AAT TTG CGT GTCAT-3′ for U6. Relative expression of miRNA was evaluated by the 2−ΔΔCq method (18). All miRNA samples and the internal reference U6 were run in a qPCR reaction. Thermal cycling conditions of the qPCR were 95°C for 10 min; 40 cycles of 95°C for 10 sec; and 40 cycles of 60°C for 60 sec.

Results

Overview of LPS and PM-induced miRNA expression profiling

To obtain a comprehensive view of the miRNA profile of BM-MSCs stimulated with LPS and PM, Illumine HiSeq 2000 technology was used to detect the global expression level annotated in Sanger miRBase 19.0. A total of 67 miRNAs demonstrated different expression patterns between the LPS and control groups (BM-MSCs without stimulation), with 32 downregulated miRNAs (Table I)and 35 upregulated miRNAs (Table II). In PM-treated cells, the downregulation (Table III) and upregulation (Table IV) were observed for 51 and 46 miRNAs, respectively. To assess the sequencing quality, the length distribution of the clean reads was summarized. Few differences were observed in the length distribution of the sequences from the control and trial libraries (Fig. 1). The most abundant sequence reads were 22, 23, 21 and 24 nt in length, which is consistent with the typical small RNA distribution of mammals.

Table I

Downregulated miRs in bone marrow-derived mesenchymal stem cells stimulated with lipopolysaccharide.

Table I

Downregulated miRs in bone marrow-derived mesenchymal stem cells stimulated with lipopolysaccharide.

miR nameFold change
hsa-let-7a-3p0.333333333
hsa-miR-10.157248157
hsa-miR-125a-5p0.403608737
hsa-miR-132-5p0.161290323
hsa-miR-1370.253012048
hsa-miR-154-3p0.153846154
hsa-miR-15a-5p0.359223301
hsa-miR-181a-5p0.287637856
hsa-miR-181c-3p0.450000000
hsa-miR-186-5p0.128352490
hsa-miR-199a-3p0.309404320
hsa-miR-199b-3p0.309404320
hsa-miR-221-3p0.280632896
hsa-miR-222-3p0.374696936
hsa-miR-24-3p0.333686441
hsa-miR-29a-3p0.187765957
hsa-miR-30d-5p0.437901499
hsa-miR-30e-5p0.300376648
hsa-miR-323a-3p0.182186235
hsa-miR-323b-3p0.094527363
hsa-miR-335-5p0.413793103
hsa-miR-337-3p0.482758621
hsa-miR-361-5p0.454545455
hsa-miR-369-3p0.404193782
hsa-miR-374a-5p0.271111111
hsa-miR-380-3p0.400000000
hsa-miR-411-3p0.083179298
hsa-miR-425-5p0.016032811
hsa-miR-450a-5p0.183206107
hsa-miR-708-5p0.258064516
hsa-miR-9410.131578947
hsa-miR-99b-5p0.476327116

[i] miR, microRNA.

Table II

Upregulated miRs in bone marrow-derived mesenchymal stem cells stimulated with lipopolysaccharide.

Table II

Upregulated miRs in bone marrow-derived mesenchymal stem cells stimulated with lipopolysaccharide.

miR nameFold change
hsa-let-7b-5p2.918778428
hsa-miR-100-5p10.847709700
hsa-miR-106a-5p2.083333333
hsa-miR-106b-5p5.405405405
hsa-miR-125a-3p3.090909091
hsa-miR-126-3p2.282608696
hsa-miR-12810.200000000
hsa-miR-136-3p2.000000000
hsa-miR-138-5p15.218750000
hsa-miR-146a-5p2.125000000
hsa-miR-155-5p19.623376620
hsa-miR-17-5p5.828767123
hsa-miR-18a-5p4.888888889
hsa-miR-199a-5p32.703703700
hsa-miR-20a-5p4.294478528
hsa-miR-218-5p4.400000000
hsa-miR-22-5p2.133333333
hsa-miR-224-5p12.050000000
hsa-miR-26a-5p8.077169132
hsa-miR-26b-5p2.234290470
hsa-miR-32-3p2.750000000
hsa-miR-34a-5p2.252148997
hsa-miR-3591-5p3.214285714
hsa-miR-361-3p4.136363636
hsa-miR-369-5p2.222222222
hsa-miR-424-3p9.666666667
hsa-miR-425-3p3.461538462
hsa-miR-455-3p6.421052632
hsa-miR-486-5p3.418918919
hsa-miR-497-5p2.230769231
hsa-miR-548d-5p2.800000000
hsa-miR-574-5p7.185185185
hsa-miR-7-5p4.600000000
hsa-miR-9412.250000000
hsa-miR-99a-5p10.116279070

[i] miR, microRNA.

Table III

Downregulated miRs in bone marrow derived mesenchymal stem cells stimulated with PAM3CSK4.

Table III

Downregulated miRs in bone marrow derived mesenchymal stem cells stimulated with PAM3CSK4.

miR nameFold change
hsa-let-7a-3p0.339080460
hsa-miR-10.090909091
hsa-miR-101-3p0.413533835
hsa-miR-103a-3p0.169556840
hsa-miR-106b-3p0.383177570
hsa-miR-1070.200000000
hsa-miR-10a-5p0.263522618
hsa-miR-10a-3p0.406250000
hsa-miR-10b-5p0.276625387
hsa-miR-125b-5p0.449112979
hsa-miR-125b-1-3p0.409638554
hsa-miR-132-5p0.322580645
hsa-miR-1340.266187050
hsa-miR-1370.409638554
hsa-miR-140-5p0.369565217
hsa-miR-146b-5p0.250539957
hsa-miR-148a-5p0.385714286
hsa-miR-148b-5p0.458333333
hsa-miR-151a-3p0.343228200
hsa-miR-154-3p0.1111111110
hsa-miR-181c-3p0.316666667
hsa-miR-186-5p0.223180077
hsa-miR-190a0.284424379
hsa-miR-191-5p0.262476895
hsa-miR-199a-3p0.255945887
hsa-miR-199b-3p0.255945887
hsa-miR-21-3p0.239819005
hsa-miR-2100.234042553
hsa-miR-221-3p0.215810131
hsa-miR-222-3p0.383072515
hsa-miR-27a-5p0.405405405
hsa-miR-27b-3p0.278292181
hsa-miR-29a-3p0.392021277
hsa-miR-30c-2-3p0.431818182
hsa-miR-30d-5p0.423982869
hsa-miR-30e-5p0.284369115
hsa-miR-323a-3p0.287449393
hsa-miR-323b-3p0.074626866
hsa-miR-335-3p0.438016529
hsa-miR-34c-5p0.138211382
hsa-miR-369-3p0.149674620
hsa-miR-374a-5p0.182222222
hsa-miR-379-5p0.346481876
hsa-miR-411-3p0.118299445
hsa-miR-425-5p0.026099925
hsa-miR-450a-5p0.114503817
hsa-miR-493-5p0.460829493
hsa-miR-671-5p0.326530612
hsa-miR-708-5p0.370967742
hsa-miR-92a-3p0.430016863
hsa-miR-99b-5p0.199426112

[i] miR, microRNA.

Table IV

Upregulated miRs in bone marrow-derived mesenchymal stem cells stimulated with PAM3CSK4.

Table IV

Upregulated miRs in bone marrow-derived mesenchymal stem cells stimulated with PAM3CSK4.

miR nameFold change
hsa-let-7a-5p4.209545028
hsa-let-7b-5p6.022742040
hsa-let-7c3.173728814
hsa-let-7d-5p11.85106383
hsa-let-7e-5p2.807106599
hsa-let-7f-5p2.787247317
hsa-let-7g-5p2.138705416
hsa-miR-100-5p9.835217133
hsa-miR-106b-5p4.783783784
hsa-miR-127-3p2.286982249
hsa-miR-1307-5p2.181818182
hsa-miR-130a-3p2.319148936
hsa-miR-136-3p2.102564103
hsa-miR-146a-5p22.854166670
hsa-miR-155-5p5.000000000
hsa-miR-15b-5p3.800000000
hsa-miR-17-3p2.000000000
hsa-miR-185-5p2.1111111110
hsa-miR-18a-5p2.222222222
hsa-miR-193b-3p4.820512821
hsa-miR-195-5p16.080000000
hsa-miR-196a-5p2.655948553
hsa-miR-214-3p84.462585030
hsa-miR-23a-3p3.709433962
hsa-miR-23b-3p5.725806452
hsa-miR-23c6.500000000
hsa-miR-24-3p29.494703390
hsa-miR-27b-5p5.000000000
hsa-miR-28-5p2.307692308
hsa-miR-320a2.166666667
hsa-miR-335-5p2.379310345
hsa-miR-374b-3p4.285714286
hsa-miR-376c-3p2.636363636
hsa-miR-377-3p2.133333333
hsa-miR-4213.884615385
hsa-miR-423-5p2.112903226
hsa-miR-424-5p2.350000000
hsa-miR-431-5p3.600000000
hsa-miR-45106.818181818
hsa-miR-4842.535714286
hsa-miR-487b3.000000000
hsa-miR-493-3p2.100000000
hsa-miR-5982.300000000
hsa-miR-92b-3p3.000000000
hsa-miR-9415.400000000
hsa-miR-99a-5p2.523255814

[i] miR, microRNA.

qPCR verification of known miRNAs

For identification of the authenticity of the miRNAs detected by Illumine HiSeq 2000 technology, 40 known miRNAs from Tables IIV were randomly selected for further validation with additional samples by qPCR. As presented in Fig. 2, the qPCR data were highly consistent with Illumine HiSeq 2000 technology, confirming the reliability of the HiSeq 2000 sequencing data.

Novel miRNA identification

To identify the novel, unannotated miRNAs in human BM-MSCs, the unlabeled reads were analyzed by miRDeep2. The present study focused on six potential novel miRNAs with the most frequent appearance among all the samples and the highest miRDeep2 score. The expression levels of these six miRNAs were further validated by qPCR. Compared with Illumine HiSeq 2000 sequencing, qPCR indicated the same trend of expression levels for the six novel miRNA candidates (Fig. 3).

Predicted target genes of LPS- and PM-responsive miRNAs

To characterize the DE miRNAs between LPS, PM and the controls, the target genes of the DE miRNAs were predicted using three different miRNA target prediction algorithms (TargetScan, MicroCosm and miRanda). Subsequently, the target genes that were identified in all three databases underwent GO analysis. In the LPS group, the targets genes of downregulated miRNAs were significantly enriched in transcripts from the RNA polymerase II promoter and included regulation of macromolecule metabolic processes, gene expression and nucleobase-containing compound metabolic processes; the most enriched GO terms of targets genes of upregulated miRNAs included anatomical structure morphogenesis, regulation of cellular processes, biological processes and cell development. In the PM group, significantly enriched GO terms of targets genes of down-regulated miRNAs included regulation of biological process, system development, transcription from RNA polymerase II promoter and cell differentiation; the most enriched GO terms of targets genes of downregulated miRNAs included regulation of signal transduction, cellular process, cell communication and biological process (Fig. 4). KEGG pathway analysis indicated that the predicted target genes were significantly enriched in a wide range of pathways. LPS-regulated miRNAs predominantly target mitogen activated protein kinases (MAPKs), estrogen, T cell receptors, phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3)-Akt, the mammalian target of rapamycin signaling pathway and cancer-associated signaling pathways, while PM-regulated miRNAs target the P13-Akt, ras-related protein 1, neurotrophin, MAPK and Ras signaling pathways (Fig. 5).

Discussion

A number of studies have reported the expression of TLRs upon MSC and TLR activation in MSCs regulates their functions in immunomodulation, migration, repair and regeneration of damaged tissues/organs (1924). In addition, a previous study reported that TLR2 agonists (PM) and/or TLR4 agonists (LPS) were able to activate MSCs, thus modulating their hematopoiesis-supporting role in vitro (10). The present study hypothesized that the miRNAs induced by TLR activation may control MSC processes, including immunomodulation, migration, and repair and regeneration of damaged tissues/organs. The present study analyzed the differential miRNA expression profiles in the MSCs derived from human fresh bone marrow and stimulated with LPS or PM. By high-throughput sequencing, compared with the expression levels in unstimulated MSCs, 67 miRNAs were identified with 35 miRNAs upregulated and 32 downregulated following LPS stimulation; 97 miRNAs with 46 miRNAs upregulated and 51 downregulated following PM stimulation. Data from the present study demonstrated that the responses to LPS or PM prompted quick and specific in vitro alterations to the miRNA expression profile of BM-MSCs.

miR-425, which has been demonstrated to be DE and the most markedly downregulated, was observed in the LPS and PM-stimulated BM-MSCs. It was reported that upregulated expression of miR-425 was associated with a variety of types of cancer, including renal and gastric cancers (25,26). A recent study demonstrated that nuclear factor-κB (NF-κB)-dependent miR-425 upregulation promotes gastric cancer cell growth by targeting phosphatase and tensin homolog in response to IL-1β treatment (25,26). Furthermore, this indicates that the action of miR-425 may mediate cell proliferation. However, to the best of our knowledge, there are no reports investigating the effects of miR-425 in BM-MSCs. Compared with previous studies, the marked difference in expression level of miR-425 indicates that miRNAs may respond differently in LPS or PM-stimulated BM-MSCs and exert different effects in the different groups.

In accordance with the miRNA results demonstrating upregulation of miR-199a in human BM-MSCs during osteoblastic differentiation and chondrogenesis (27), the most abundantly expressed miRNA in LPS-stimulated BM-MSCs was miR-199a, which was also upregulated in LPS-stimulated leukocytes derived from cord blood (CB) (28). Laine et al (27) demonstrated that cells transfected with pre-miR-199a reduced the proliferation of human MSCs and miR-199a restricts chondrogenic differentiation by suppressing the expression of SRY-box 9, which is a positive regulator of chondrogenesis (29). In addition, miR-199a has been identified as a negative regulator of chondrogenesis (30). Wu et al (31) inferred that the overexpression of miR-199a-5p inhibits the proliferation of keloid fibroblasts. However, Shi et al (32) demonstrated that overexpression of miR-199a-5p promoted the proliferation of porcine preadipocytes and suppressed adipogenic differentiation. These findings indicate that increased expression of miR-199a in LPS-stimulated BM-MSCs may be a key regulator for MSC differentiation and proliferation. As miR-199 was observed to exert opposing effects on proliferation in different cells, this is likely explained by miRNA-mRNA targeting, and the different interactions among tissue and cell types and physiological/pathological conditions (33). Further studies are required to investigate the exact underlying mechanism of miR-199a action in BM-MSCs.

The miRNA that was most abundantly expressed in human PM-stimulated BM-MSCs was miR-214. Previous studies reported that miR-214 may be important in promoting myoblast proliferation and differentiation (34); overexpression of miR-214 was connected to gastric cancer tissue, and deletion of miR-214 inhibited the proliferation, migration and invasion of gastric cancer cells (35). The results are consistent with recent findings by Zhang et al (36), that miR-214 is overexpressed in nasopharyngeal carcinoma tissues and cell lines, and that knockdown of miR-214 suppressed cell proliferation and induced apoptosis. However, Derfoul et al (37), demonstrated that overexpression of miR-214 inhibited the proliferation and invasion of breast cancer cells. Thus, the abnormal expression of miR-214 may be closely associated with certain types of cancer by affecting cell growth, differentiation and migration.

However, whether the TLR signaling pathway is regulated by the above-mentioned miRNAs, which are expressed most differentially, and whether their target genes are involved in TLR signal transduction, requires further investigation.

To investigate the function of the LPS- or PM-induced DE miRNAs further, the target genes were predicted, and GO and KEGG pathway analyses were applied to these predicted target genes. The GO term annotation suggests that the presumed target genes of these DE miRNAs in BM-MSCs are involved in a broad range of physiological processes in response to stimuli. Consistent with the GO assay, among those pathways, the MAPK and PI3-Akt signaling pathway are notable, as they have been reported to be important in promoting cell proliferation, growth and differentiation (38).

Notably, in the present study, a number of the verified TLR signaling pathway-associated miRNAs were markedly DE between the control and trial groups. For example, in the TLR4 agonist group, the associated markedly DE miRNAs were miR-146a, miR-155, miR-132, miR-29, miR-199a-5p, let-7b, miR-24, miR-221, miR-181, miR-106a/b, miR-20a, miR-26a and miR-34a (14,15,39-48). In the TLR2 agonist group, the associated markedly DE miRNAs were miR-146a/b, miR-155, miR-132, miR-29, miR-199a-3p, let-7b, miR-24, miR-221, miR-181, miR-106b, miR-92a, miR-210, miR-27b, miR-21, miR-125b, miR-101 and miR-148a/b (12,15,16,38-45,49-54). These miRNAs in BM-MSCs may suggest a potentially important role in TLR signaling regulation. Furthermore, miR-146b was observed to be upregulated in CB leukocytes following LPS stimulation and may control LPS-stimulated neonatal early phases of inflammation via negative feedback loops (28). Ma et al (55) reported that miR-301a was upregulated in umbilical cord-MSCs stimulated with LPS, while miR-301a was not regulated by LPS and PM in the dataset produced in the present study, this notable difference is likely due to the different experimental methods and sources of material.

TLR activation triggers myeloid differentiation primary response gene 88-dependent and -independent downstream signaling cascades, leading to the activation of a number of transcription factors and genes (5661). These downstream signaling molecules predominantly include transforming growth factor-β-activated kinase, IL-1R-associated kinases, MAPKs, NF-κB and tumor necrosis factor-receptor-associated factor 6 (59). It has been demonstrated that following stimulation with ligands specific to TLRs, the NF-κB, MAPK, and PI3K signaling pathways were activated with a subsequent induction of multiple genes and cytokines (62). Furthermore, combined with KEGG pathways analysis, the results of the present study indicate a critical role for miRNAs, via the TLR signaling pathway, in the growth, differentiation, and migration of LPS and PM-induced BM-MSCs.

During comparison of these two sets of experiments, a number of the TLR signaling pathway-associated miRNAs (miR-146a, miR-155, let-7b, miR-106b, miR-132, miR-29, miR-221 and miR-181) were found to be the same, and the up- or downregulation of specific miRNAs was also consistent, suggesting that LPS- or PM-induced miRNAs may have common targets and exert similar effects in BM-MSCs. Future studies are required to validate these predictions.

Six novel miRNAs that demonstrated marked changes in response to TLR activation were observed in the present study, which were not similar to any known miRNAs. Future investigations are required to predict and functionally validate these DE miRNA targets.

In conclusion, the results of the current study identify the global expression change of miRNAs in BM-MSCs stimulated with LPS and PM, providing a framework for further analysis of miRNAs and their role in mediating TLR signals to regulate the functions of BM-MSCs.

Acknowledgments

The authors would like to thank Dr Jun Liu for reviewing the manuscript. The present study was supported by the National Natural Science Foundation (grant no. 81270573) and the Natural Science Foundation of Anhui Higher Education Institutions (grant no. KJ2012Z188).

Abbreviations:

BM-MSCs

bone marrow-derived mesenchymal stromal cells

GO

gene ontology

LPS

lipopolysaccharides

MAPKs

mitogen-activated protein kinases

miRNAs

microRNAs

NF-κB

nuclear factor-κB

RT-qPCR

reverse transcription-quantitative polymerase chain reaction

TLRs

toll-like receptors

References

1 

Yates LA, Norbury CJ and Gilbert RJ: The long and short of microRNA. Cell. 153:516–519. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S and Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 284:143–147. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, et al: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 418:41–49. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Charbord P: Bone marrow mesenchymal stem cells: Historical overview and concepts. Hum Gene Ther. 21:1045–1056. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Oreffo RO, Cooper C, Mason C and Clements M: Mesenchymal stem cells: Lineage, plasticity and skeletal therapeutic potential. Stem Cell Rev. 1:169–178. 2005. View Article : Google Scholar

8 

Hwa Cho H, Bae YC and Jung JS: Role of toll-like receptors on human adipose-derived stromal cells. Stem Cells. 24:2744–2752. 2006. View Article : Google Scholar : PubMed/NCBI

9 

van den Berk LC, Jansen BJ, Siebers-Vermeulen KG, Netea MG, Latuhihin T, Bergevoet S, Raymakers RA, Kögler G, Figdor CC, Adema GJ and Torensma R: Toll-like receptor triggering in cord blood mesenchymal stem cells. J Cell Mol Med. 13:3415–3426. 2009. View Article : Google Scholar

10 

Wang X, Cheng Q, Li L, Wang J, Xia L, Xu X and Sun Z: Toll-like receptors 2 and 4 mediate the capacity of mesenchymal stromal cells to support the proliferation and differentiation of CD34+cells. Exp Cell Re. 318:196–206. 2012. View Article : Google Scholar

11 

Benakanakere MR, Li Q, Eskan MA, Singh AV, Zhao J, Galicia JC, Stathopoulou P, Knudsen TB and Kinane DF: Modulation of TLR2 protein expression by miR-105 in human oral keratinocytes. J Biol Chem. 284:23107–23115. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Quinn EM, Wang JH, O'Callaghan G and Redmond HP: MicroRNA-146a is upregulated by and negatively regulates TLR2 signaling. PLoS One. 8:e622322013. View Article : Google Scholar : PubMed/NCBI

13 

O'Hara SP, Splinter PL, Gajdos GB, Trussoni CE, Fernandez-Zapico ME, Chen XM and LaRusso NF: NFkappaB p50-CCAAT/enhancer-binding protein beta (C/EBPbeta)-mediated transcriptional repression of microRNA let-7i following microbial infection. J Biol Chem. 285:216–225. 2010. View Article : Google Scholar :

14 

Yang K, He YS, Wang XQ, Lu L, Chen QJ, Liu J, Sun Z and Shen WF: MiR-146a inhibits oxidized low-density lipoprotein-induced lipid accumulation and inflammatory response via targeting toll-like receptor 4. FEBS Lett. 585:854–860. 2011. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Curtale G, Mirolo M, Renzi TA, Rossato M, Bazzoni F and Locati M: Negative regulation of Toll-like receptor 4 signaling by IL-10-dependent microRNA-146b. Proc Natl Acad Sci USA. 110:11499–11504. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Ceppi M, Pereira PM, Dunand-Sauthier I, Barras E, Reith W, Santos MA and Pierre P: MicroRNA-155 modulates the interleukin-1 signaling pathway in activated human monocyte-derived dendritic cells. Proc Natl Acad Sci USA. 106:2735–2740. 2009. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Hwa Cho H, Bae YC and Jung JS: Role of toll-like receptors on human adipose-derived cells. Stem Cells. 24:2744–2752. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Pevsner-Fischer M, Morad V, Cohen-Sfady M, Rousso-Noori L, Zanin-Zhorov A, Cohen S, Cohen IR and Zipori D: Toll-like receptors and their ligands control mesenchymal stem cell functions. Blood. 109:1422–1432. 2007. View Article : Google Scholar

21 

Liotta F, Angeli R, Cosmi L, Filì L, Manuelli C, Frosali F, Mazzinghi B, Maggi L, Pasini A, Lisi V, et al: Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T cell modulatory activity by 634 impairing Notch signaling. Stem Cells. 26:279–289. 2008. View Article : Google Scholar

22 

Opitz CA, Litzenburger UM, Lutz C, Lanz TV, Tritschler I, Köppel A, Tolosa E, Hoberg M, Anderl J, Aicher WK, et al: Toll-like receptor engagement enhances the immunosuppressive properties of human bone marrow-derived mesenchymal stem cells by inducing indoleamine-2,3-dioxygenase-1 via interferon-beta and protein kinase R. Stem Cells. 27:909–919. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Tomchuck SL, Zwezdaryk KJ, Coffelt SB, Waterman RS, Danka ES and Scandurro AB: Toll-like receptors on human mesenchymal stem cells drive their migration and immunomodulating responses. Stem Cells. 26:99–107. 2008. View Article : Google Scholar

24 

Romieu-Mourez R, François M, Boivin MN, Bouchentouf M, Spaner DE and Galipeau J: Cytokine modulation of TLR expression and activation in mesenchymal stromal cells leads to a proinflammatory phenotype. J Immunol. 182:7963–7973. 2009. View Article : Google Scholar : PubMed/NCBI

25 

White NM, Bao TT, Grigull J, Youssef YM, Girgis A, Diamandis M, Fatoohi E, Metias M, Honey RJ, Stewart R, et al: miRNA profiling for clear cell renal cell carcinoma: Biomarker discovery and identification of potential controls and consequences of miRNA dysregulation. J Urol. 186:1077–1083. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Ma J, Liu J, Wang Z, Gu X, Fan Y, Zhang W, Xu L, Zhang J and Cai D: NF-kappaB-dependent MicroRNA-425 upregulation promotes gastric cancer cell growth by targeting PTEN upon IL-1β induction. Mol Cancer. 13:402014. View Article : Google Scholar

27 

Laine SK, Alm JJ, Virtanen SP, Aro HT and Laitala-Leinonen TK: MicroRNAs miR-96, miR-124 and miR-199a regulate gene expression in human bone marrow-derived mesenchymal stem cells. J Cell Biochem. 113:2687–2695. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Chen J, Liu Z and Yang Y: In vitro screening of LPS-induced miRNAs in leukocytes derived from cord blood and their possible roles in regulating TLR signals. Pediatr Res. 75:595–602. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Akiyama H: Control of chondrogenesis by the transcription factor Sox9. Mod Rheumatol. 18:213–219. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Lin EA, Kong L, Bai XH, Luan Y and Liu CJ: miR-199a, a bone morphogenic protein 2-responsive MicroRNA, regulates chondrogenesis via direct targeting to Smad1. J Biol Chem. 284:11326–11335. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Wu ZY, Lu L, Liang J, Guo XR, Zhang PH and Luo SJ: Keloid microRNA expression analysis and the influence of miR-199a-5p on the proliferation of keloid fibroblasts. Genet Mol Res. 13:2727–2738. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Shi XE, Li YF, Jia L, Ji HL, Song ZY, Cheng J, Wu GF, Song CC, Zhang QL, Zhu JY and Yang GS: MicroRNA-199a-5p affects porcine preadipocyte proliferation and differentiation. Int J Mol Sci. 15:8526–8538. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M and Rajewsky N: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Feng Y, Cao JH, Li XY and Zhao SH: Inhibition of miR-214 expression represses proliferation and differentiation of C2C12 myoblasts. Cell Biochem Funct. 29:378–383. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Yang TS, Yang XH, Wang XD, Wang YL, Zhou B and Song ZS: MiR-214 regulate gastric cancer cell proliferation, migration and invasion by targeting PTEN. Cancer Cell Int. 13:682013. View Article : Google Scholar : PubMed/NCBI

36 

Zhang ZC, Li YY, Wang HY, Fu S, Wang XP, Zeng MS, Zeng YX and Shao JY: Knockdown of miR-214 promotes apoptosis and inhibits cell proliferation in nasopharyngeal carcinoma. PLoS One. 9:e861492014. View Article : Google Scholar : PubMed/NCBI

37 

Derfoul A, Juan AH, Difilippantonio MJ, Palanisamy N, Ried T and Sartorelli V: Decreased microRNA-214 levels in breast cancer cells coincides with increased cell proliferation, invasion and accumulation of the Polycomb Ezh2 methyltransferase. Carcinogenesis. 32:1607–1614. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Brzezianska E and Pastuszak-Lewandoska D: A minireview: The role of MAPK/ERK and PI3K/Akt pathways in thyroid follicular cell-derived neoplasm. Front Biosci (Landmark Ed). 16:422–439. 2011. View Article : Google Scholar

39 

Nahid MA, Yao B, Dominguez-Gutierrez PR, Kesavalu L, Satoh M and Chan EK: Regulation of TLR2-mediated tolerance and cross-tolerance through IRAK4 modulation by miR-132 and miR-212. J Immunol. 190:1250–1263. 2013. View Article : Google Scholar :

40 

Ahmed F, Shiraishi T, Vessella RL and Kulkarni P: Tumor necrosis factor receptor associated factor-4: An adapter protein overexpressed in metastatic prostate cancer is regulated by microRNA-29a. Oncol Rep. 30:2963–2968. 2013.PubMed/NCBI

41 

Chen R, Alvero AB, Silasi DA, Kelly MG, Fest S, Visintin I, Leiser A, Schwartz PE, Rutherford T and Mor G: Regulation of IKKB by miR-199a affects NF-kappB activity in ovarian cancer cells. Oncogene. 27:4712–4723. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Zhou R, O'Hara SP and Chen XM: MicroRNA regulation of innate immune responsesin epithelial cells. Cell Mol Immunol. 8:371–379. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Huang Z, Chen X, Yu B and Chen D: Cloning and functional characterization of rat stimulator of interferon genes (STING) regulated by miR-24. Dev Comp Immunol. 37:414–420. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Lai NS, Yu HC, Chen HC, Yu CL, Huang HB and Lu MC: Aberrant expression of microRNAs in T cells from patients with ankylosing spondylitis contributes to the immunopathogenesis. Clin Exp Immunol. 173:47–57. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Zhao J, Gong AY, Zhou R, Liu J, Eischeid AN and Chen XM: Downregulation of PCAF by miR-181a/b provides feedback regulation to TNF-α-induced transcription of proinflammatory genes in liver epithelial cells. J Immunol. 188:1266–1274. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Sharma A, Kumar M, Aich J, Hariharan M, Brahmachari SK, Agrawal A and Ghosh B: Posttranscriptional regulation of interleukin-10 expression by hsa-miR-106a. Proc Natl Acad Sci USA. 106:5761–5766. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Zhang M, Liu Q, Mi S, Liang X, Zhang Z, Su X, Liu J, Chen Y, Wang M, Zhang Y, et al: Both miR-17-5p and miR-20a alleviate suppressive potential of myeloid-derived suppressor cells by modulating STAT3 expression. J Immunol. 186:4716–4724. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Witwer KW, Sisk JM, Gama L and Clements JE: MicroRNA regulation of IFN-beta protein expression: Rapid and sensitive modulation of the innate immune response. J Immunol. 184:2369–2376. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Lai L, Song Y, Liu Y, Chen Q, Han Q, Chen W, Pan T, Zhang Y, Cao X and Wang Q: MicroRNA-92a negatively regulates Toll-like receptor (TLR)-triggered inflammatory response in macrophages by targeting MKK4 kinase. J Biol Chem. 288:7956–7967. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Qi J, Qiao Y, Wang P, Li S, Zhao W and Gao C: MicroRNA-210 negatively regulates LPS-induced production of proinflammatory cytokines by targeting NF-κB1 in murine macrophages. FEBS Lett. 586:1201–1207. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Jennewein C, von Knethen A, Schmid T and Brüne B: MicroRNA-27b contributes to lipopolysaccharide-mediated peroxisome proliferator-activated receptor gamma (PPARgamma) mRNA destabilization. J Biol Chem. 285:11846–11853. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Lu TX, Munitz A and Rothenberg ME: MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression. J Immunol. 182:4994–5002. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Zhu QY, Liu Q, Chen JX, Lan K and Ge BX: MicroRNA-101 targets MAPK phosphatase-1 to regulate the activation of MAPKs in macrophages. J Immunol. 185:7435–7442. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Liu X, Zhan Z, Xu L, Ma F, Li D, Guo Z, Li N and Cao X: MicroRNA-148/152 impair innate response and antigen presentation of TLR-triggered dendritic cells by targeting CaMKIIα. J Immunol. 185:7244–7251. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Ma F, Chen D, Chi Y, Chen F, Li X and Han Z: The expression and role of miR-301a in human umbilical cord-derived mesenchymal stromal cells. Cytotherapy. 15:1511–1516. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Akira S, Uematsu S and Takeuchi O: Pathogen recognition and innate immunity. Cell. 124:783–801. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Ozinsky A, Underhill DM, Fontenot JD, Hajjar AM, Smith KD, Wilson CB, Schroeder L and Aderem A: The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between toll-like receptors. Proc Natl Acad Sci USA. 97:13766–13771. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Pasare C and Medzhitov R: Toll-like receptors: Linking innate and adaptive immunity. Adv Exp Med Biol. 560:11–18. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Hoebe K, Jiang Z, Georgel P, Tabeta K, Janssen E, Du X and Beutler B: TLR signaling pathways: Opportunities for activation and blockade in pursuit of therapy. Curr Pharm Des. 12:4123–4134. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Kawai T and Akira S: TLR signaling. Semin Immunol. 19:24–32. 2007. View Article : Google Scholar : PubMed/NCBI

61 

O'Neill LA and Bowie AG: The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol. 7:353–364. 2007. View Article : Google Scholar : PubMed/NCBI

62 

DelaRosa O and Lombardo E: Modulation of adult mesenchymal stem cells activity by toll-like receptors: Implications on therapeutic potential. Mediators Inflamm. 2010:8656012010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 13 Issue 6

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
Wang X, Zhu Y, Xu B, Wang J and Liu X: Identification of TLR2 and TLR4‑induced microRNAs in human mesenchymal stem cells and their possible roles in regulating TLR signals. Mol Med Rep 13: 4969-4980, 2016
APA
Wang, X., Zhu, Y., Xu, B., Wang, J., & Liu, X. (2016). Identification of TLR2 and TLR4‑induced microRNAs in human mesenchymal stem cells and their possible roles in regulating TLR signals. Molecular Medicine Reports, 13, 4969-4980. https://doi.org/10.3892/mmr.2016.5197
MLA
Wang, X., Zhu, Y., Xu, B., Wang, J., Liu, X."Identification of TLR2 and TLR4‑induced microRNAs in human mesenchymal stem cells and their possible roles in regulating TLR signals". Molecular Medicine Reports 13.6 (2016): 4969-4980.
Chicago
Wang, X., Zhu, Y., Xu, B., Wang, J., Liu, X."Identification of TLR2 and TLR4‑induced microRNAs in human mesenchymal stem cells and their possible roles in regulating TLR signals". Molecular Medicine Reports 13, no. 6 (2016): 4969-4980. https://doi.org/10.3892/mmr.2016.5197