Open Access

Comprehensive bioinformatics analysis of critical lncRNAs, mRNAs and miRNAs in non‑alcoholic fatty liver disease

  • Authors:
    • Huiling Wu
    • Xi Song
    • Yuntao Ling
    • Jin Zhou
    • Zhen Tao
    • Yuying Shen
  • View Affiliations

  • Published online on: February 5, 2019     https://doi.org/10.3892/mmr.2019.9931
  • Pages: 2649-2659
  • Copyright: © Wu 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

Non‑alcoholic fatty liver disease (NAFLD) is the most common fatty liver disease in developed countries, in which fat accumulation in the liver is induced by non‑alcoholic factors. The present study was conducted to identify NAFLD‑associated long non‑coding RNAs (lncRNAs), mRNAs and microRNAs (miRNAs). The microarray dataset GSE72756, which included 5 NAFLD liver tissues and 5 controls, was acquired from the Gene Expression Omnibus database. Differentially expressed lncRNAs (DE‑lncRNAs) and mRNAs (DE‑mRNAs) were detected using the pheatmap package. Using the clusterProfiler package and Cytoscape software, enrichment and protein‑protein interaction (PPI) network analyses were conducted to evaluate the DE‑mRNAs. Next, the miRNA‑lncRNA‑mRNA interaction network was visualized using Cytoscape software. Additionally, RP11‑279F6.1 and AC004540.4 expression levels were analyzed by reverse transcription quantitative polymerase chain reaction. There were 318 DE‑lncRNAs and 609 DE‑mRNAs identified in the NAFLD tissues compared with the normal tissues. Jun proto‑oncogene, AP‑1 transcription factor subunit (JUN), which is regulated by AC004540.4 and RP11‑279F6.1, exhibited higher degree compared with other nodes in the PPI network. Furthermore, miR‑409‑3p and miR‑139 (targeting JUN) were predicted as PPI network nodes. In the miRNA‑lncRNA‑mRNA network, miR‑20a and B‑cell lymphoma 2‑like 11 (BCL2L11) were among the top 10 nodes. Additionally, BCL2L11, AC004540.4 and RP11‑279F6.1 were targeted by miR‑20a, miR‑409‑3p and miR‑139 in the miRNA‑lncRNA‑mRNA network, respectively. RP11‑279F6.1 and AC004540.4 expression was markedly enhanced in NAFLD liver tissues. These key RNAs may be involved in the pathogenic mechanisms of NAFLD.

Introduction

As the most frequently diagnosed fatty liver disease in developed countries, non-alcoholic fatty liver disease (NAFLD) occurs when fat is enriched in the liver due to non-alcoholic factors (1,2). The risk factors for NAFLD include metabolic syndromes, for example combined hyperlipidemia, obesity, high blood pressure and type II diabetes mellitus and insulin resistance (3,4). Non-alcoholic steatohepatitis (NASH), which is the most severe type of NAFLD, is considered to be the primary cause of cirrhosis (4). The incidence of NAFLD is 9–36.9% globally (5,6), and ~20% of people in the United States of America (75–100 million people) are affected by the disease (7). Therefore, exploring the pathogenic mechanisms of NAFLD and developing novel treatment protocols are necessary.

Silencing of fatty acid transport protein 5 (FATP5) may reverse NAFLD; therefore, the activity of hepatic FATP5 is considered critical for maintaining fatty acid flux and caloric uptake during high-fat feeding (8). Patatin-like phospholipase domain containing 3 is an NAFLD-associated gene, which is closely associated with metabolic changes in hepatocytes and lipogenesis (9,10). Interleukin-17 is associated with the proinflammatory response and hepatic steatosis in NAFLD and contributes to the steatosis-steatohepatitis transition (11). Several microRNAs (miRNAs), including miR-21, miR-122, miR-451 and miR-34a, are overexpressed in patients with NAFLD; in particular, miR-122 levels are associated with the grades of fatty liver disease and are a potential marker of NAFLD (12). By inhibiting the expression of 3-hydroxy-3-methylglutaryl-co-enzyme A reductase, miR-21 was demonstrated to mediate cholesterol and triglyceride metabolism in an NAFLD model and may be a promising diagnostic and therapeutic marker for the disease (13). miR-34a/Sirtuin 1/tumor protein p53 (p53) signaling, which is correlated with liver cell apoptosis, is inhibited by ursodeoxycholic acid and activated upon the aggravation of illness in NAFLD (14). Despite these studies, the molecular mechanisms of NAFLD are not fully understood.

In 2015, Sun et al (15) investigated the role of long non-coding RNAs (lncRNAs) in NAFLD through microarray data analysis and identified that several differentially expressed lncRNAs (DE-lncRNAs) function in the pathogenesis of NAFLD. However, the molecular regulatory mechanisms in NAFLD have not been explored in detail. Based on the expression profiles deposited by Sun et al (15), the present study additionally identified the DE-lncRNAs and differentially expressed mRNAs (DE-mRNAs) between NAFLD and normal liver tissues. In addition, key mRNAs, lncRNAs and miRNAs involved in NAFLD were also identified through protein-protein interaction (PPI) network, enrichment and miRNA-lncRNA-mRNA interaction network analyses. The expression of key RNAs were detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR).

Materials and methods

Data source

The normalized expression data and annotation data from the GSE72756 dataset were acquired from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/), which was generated using a GPL16956 Agilent-045997 Arraystar human lncRNA microarray V3 (Probe Name Version) platform. The sample set of GSE72756 included 5 NAFLD liver tissues (3 females and 2 males; mean age=38.8 years) and 5 normal liver tissues (3 females and 2 males; mean age=39.2 years). Samples used in this dataset were sourced from patients with NAFLD without other metabolic complications that were hospitalized in The Third Xiangya Hospital (Changsha, China) from March 2014 to November 2014, and NAFLD was confirmed independently by two senior pathologists by pathological examination. Liver tissues (50–100 mg) were isolated from the patients and then rapidly frozen in liquid nitrogen. Sun et al (15) deposited the microarray dataset GSE72756, and the study was approved by the Ethics Committee of the Third Xiangya Hospital of Central South University. Informed consent was obtained from all patients.

Differential expression analysis

Based on the normalized probe expression data, the R package in Linear Models for Microarray Data (16) (http://www.bioconductor.org/packages/2.9/bioc/html/limma.html) was utilized to identify and annotate the differentially expressed probes between NAFLD and normal liver tissues. A false discovery rate (FDR; adjusted P-value) <0.01 and |log fold-change (FC)|>1 were set as thresholds. Using the R package pheatmap (17) (http://cran.r-project.org/web/packages/pheatmap/index.html), a clustering heatmap was drawn for differentially expressed probes. According to the annotation information, the differentially expressed probes were divided into DE-lncRNAs and DE-mRNAs.

Enrichment analysis and PPI network analysis of DE-mRNAs

Using the R package clusterProfiler (18) (http://bioconductor.org/packages/release/bioc/html/clusterProfiler.html), Gene Ontology (GO; http://www.geneontology.org) (19) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.ad.jp/kegg) (20) pathway enrichment analyses were performed for the DE-mRNAs. The terms with FDR <0.05 were considered to be significant. Based on the Search Tool for the Retrieval of Interacting Genes (http://www.string-db.org/) (21) database, the interaction pairs among the DE-mRNAs were predicted with the threshold of required confidence >0.4. The PPI network was constructed using Cytoscape software (version 3.0.1, http://www.cytoscape.org) (22). Using the CytoNCA plugin (http://apps.cytoscape.org/apps/cytonca) (23) in Cytoscape, Closeness centrality (CC) (24), Degree Centrality (DC) (25), and Betweenness centrality (BC) (26) scores were calculated. The nodes with increased CC, DC and BC scores compared with other nodes were identified as hub nodes (27) in the PPI network.

Construction of the lncRNA-mRNA regulatory network and functional prediction of DE-lncRNAs

Based on the Pearson product-moment correlation coefficient (28), the target genes of upregulated and downregulated lncRNAs were predicted using a threshold of FDR <0.05 and correlation coefficient >0.995. Next, Cytoscape software (22) was utilized to draw lncRNA-mRNA regulatory networks. Additionally, enrichment analysis was conducted for the target genes of each DE-lncRNA using FDR <0.05 as a threshold. Using the R package clusterProfiler (18), the enriched pathways were compared to identify the significant pathways of target genes for each DE-lncRNA. An FDR <0.05 was used as the cut-off criterion.

Construction of miRNA-lncRNA-mRNA interaction network

Combined with the WEB-based gene set analysis toolkit (Webgestalt; http://www.webgestalt.org) (29), the miRNAs targeting DE-mRNAs involved in the PPI network were predicted. A count (number of target genes) ≥4 and FDR <0.05 were set as the thresholds. According to the annotation information of the DE-lncRNAs, their corresponding sequences were extracted from human reference genome hg19 (30). The mature sequences of the predicted miRNAs were downloaded from the miRbase database (http://www.mirbase.org/) (31). Using MiRanda (http://www.microrna.org) (32) and RNAhybrid (http://bibiserv.techfak.uni-bielefeld.de/rnahybrid/) (33) software, lncRNAs containing significant binding sites for the aforementioned miRNAs were predicted. The predicted results of the miRanda and RNAhybrid analyses were compared to obtain the intersecting miRNA-lncRNA pairs. Based on the obtained miRNA-lncRNA and miRNA-mRNA pairs, and the associated lncRNA-mRNA and mRNA-mRNA pairs, an miRNA-lncRNA-mRNA interaction network was constructed and subjected to topological property analysis using Cytoscape software.

Sample information and RT-qPCR

A total of 2 normal liver tissues (2 females; age range: 47–60 years; mean age=53.5 years; obtained from March to June 2017 via surgical resection) and 2 NAFLD liver tissues (2 males; age range: 41–47 years; mean age=44 years; obtained from April to May 2017 via surgical resection) were provided by Nanjing First Hospital (Nanjing, China). The present study was approved by the Ethics Committee of Nanjing First Hospital. Informed consent was obtained from all patients.

Using TRIzol® reagent (Thermo Fisher Scientific, Inc., Waltham, MA, USA), total RNA was extracted from the samples. The PrimeScript RT Master MIX kit (Takara Bio, Inc., Otsu, Japan) was used to synthesize first-strand cDNA. RT-qPCR was performed using Power SYBR Green PCR Master Mix (cat. no., 4367659; Thermo Fisher Scientific, Inc.) on an ABI 7500 FAST real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The thermocycler conditions for qPCR were as follows: Initial denaturation (50°C, 3 min); 40 cycles of denaturation (95°C, 3 min), annealing (95°C, 10 sec), and extension (60°C, 30 sec). The specificity of the primer amplicons was examined by melting curve analysis. The comparative Cq method (34) was employed to quantify target mRNA and miRNA expression. mRNA expression was normalized to that of GAPDH. The primers used in the present study were as follows: GAPDH-forward (F)-5′-TGACAACTTTGGTATCGTGGAAGG-3′; GAPDH-reverse (R)-5′-AGGCAGGGATGATGTTCTGGAGAG-3′; RP11-279F6.1-h-F-5′-CGGACATAGCCAACGCACCT-3′; RP11-279F6.1-R-5′-TTCATACTTCTGCTGCGTCCA-3′; AC004540.4-F-5′-TTCACAACACACTCAAAGCCT-3′; AC004540.4-R-5′-CAACTGCACTCCAAATGGCTA-3′.

Statistical analysis

All data are expressed as the mean ± standard error of the mean, and the differences between the two groups were compared by Student's t-test. Statistical analyses were performed using SPSS 22.0 software (IBM Corp., Armonk, NY, USA), and GraphPad Prism 5 (GraphPad Software, Inc., La Jolla, CA, USA) was used to visualize the results. P<0.05 was considered to indicate a statistically significant difference.

Results

Differential expression analysis

The clustering heatmap for differentially expressed probes is presented in the Fig. 1. There were 318 DE-lncRNAs, including 105 upregulated and 213 downregulated, and 609 DE-mRNAs, including 353 upregulated and 256 downregulated, in the NAFLD liver tissues compared with the normal liver tissues.

Enrichment analysis and PPI network analysis of DE-mRNAs

The top 5 GO ‘biological process’ (BP), GO ‘cellular component’ (CC), GO ‘molecular function’ (MF) and KEGG terms enriched in the DE-mRNAs analysis are summarized in Table I. For the upregulated mRNAs, the enriched GO and KEGG terms primarily included ‘carboxylic acid metabolic process’ (GO_BP; FDR=1.24×10−20), ‘cytoplasmic part’ (GO_CC; FDR=8.66×10−12), ‘molecular function’ (GO_MF; FDR=1.73×10−14), and ‘metabolic pathways’ (KEGG pathway; FDR=2.65×10−09). Downregulated mRNAs were primarily enriched in ‘single-organism process’ (GO_BP; FDR=1.52×10−12), ‘extracellular space’ (GO_CC; FDR=5.95×10−11), ‘molecular function’ (GO_MF; FDR=6.89×10−09), and ‘adrenergic signaling in cardiomyocytes’ (KEGG pathway; FDR=4.34×10−2). The PPI network for the DE-mRNAs involved 442 nodes and 1,409 edges, and it was identified that Jun proto-oncogene, AP-1 transcription factor subunit (JUN) interacted with B-cell lymphoma 2 (Bcl-2)-like 11 (BCL2L11), as demonstrated in Fig. 2. The top 15 nodes, including JUN, with the highest BC, CC and DC scores are summarized in Table II.

Table I.

Top 5 functions and pathways enriched in the analysis of differentially expressed mRNAs.

Table I.

Top 5 functions and pathways enriched in the analysis of differentially expressed mRNAs.

CategoryTermIDDescriptionP-valueFDRCount
UpregulatedGO_BPGO:0019752Carboxylic acid metabolic process 2.90×10−24 1.24×10−20  69
GO_BPGO:0032787Monocarboxylic acid metabolic process 7.56×10−24 1.61×10−20  53
GO_BPGO:0006082Organic acid metabolic process 1.14×10−23 1.62×10−20  73
GO_BPGO:0043436Oxoacid metabolic process 2.13×10−23 2.27×10−20  72
GO_BPGO:0044281Small molecule metabolic process 7.36×10−22 6.27×10−19110
GO_CCGO:0044444Cytoplasmic part 1.98×10−14 8.66×10−12206
GO_CCGO:0044432Endoplasmic reticulum part 1.50×10−12 3.27×10−10  55
GO_CCGO:0005783Endoplasmic reticulum 3.69×10−12 5.38×10−10  68
GO_CCGO:0044421Extracellular region part 5.42×10−12 5.93×10−10120
GO_CCGO:0005789Endoplasmic reticulum membrane 1.20×10−11 1.05×10−9  48
GO_MFGO:0003674Molecular function 2.54×10−17 1.73×10−14324
GO_MFGO:0003824Catalytic activity 1.18×10−15 4.01×10−13163
GO_MFGO:0048037Cofactor binding 4.10×10−12 9.30×10−10  25
GO_MFGO:0016491Oxidoreductase activity 8.89×10−12 1.51×10−9  41
GO_MFGO:0050662Coenzyme binding 2.34×10−8 3.18×10−6  17
KEGG Pathwayhsa01100Metabolic pathways 1.02×10−11 2.65×10−9  72
KEGG Pathwayhsa00040Pentose and glucuronate interconversions 2.24×10−5 2.59×10−3  7
KEGG Pathwayhsa01200Carbon metabolism 3.94×10−5 2.59×10−3  12
KEGG Pathwayhsa00830Retinol metabolism 4.32×10−5 2.59×10−3     9
KEGG Pathwayhsa05204Chemical carcinogenesis 4.99×10−5 2.59×10−3  10
DownregulatedGO_BPGO:0044699Single-organism process 4.23×10−16 1.52×10−12197
GO_BPGO:0032501Multicellular organismal process 1.48×10−12 1.85×10−9128
GO_BPGO:0044707 Single-multicellular organism process 1.54×10−12 1.85×10−9125
GO_BPGO:0008150Biological process 6.60×10−12 5.93×10−9210
GO_BPGO:0051128Regulation of cellular component organization 1.24×10−11 8.92×10−9  58
GO_CCGO:0005615Extracellular space 1.54×10−13 5.95×10−11  52
GO_CCGO:0044449Contractile fiber part 1.04×10−11 2.02×10−9  19
GO_CCGO:0005576Extracellular region 2.82×10−11 3.64×10−9103
GO_CCGO:0043292Contractile fiber 4.73×10−11 4.58×10−9  19
GO_CCGO:0030017Sarcomere 1.73×10−10 1.34×10−8  17
GO_MFGO:0003674Molecular function 1.33×10−11 6.89×10−9213
GO_MFGO:0005515Protein binding 8.48×10−9 2.19×10−6159
GO_MFGO:0008307Structural constituent of muscle 1.73×10−8 2.99×10−6     8
GO_MFGO:0008092Cytoskeletal protein binding 3.61×10−8 4.67×10−6  29
GO_MFGO:0005488Binding 1.29×10−6 1.34×10−6187
Pathwayhsa04261Adrenergic signaling in cardiomyocytes 2.73×10−44.34×10°     8
Pathwayhsa00512Mucin type O-glycan biosynthesis 4.43×10−44.34×10°     4

[i] GO, Gene Ontology; BP, biological process; CC, cell component; MF, molecular function; KEGG, Kyoto Encyclopedia of Genes and Genomes; FDR, false discovery rate.

Table II.

Top 15 nodes with increased DC, BC and CC scores in the protein-protein interaction network.

Table II.

Top 15 nodes with increased DC, BC and CC scores in the protein-protein interaction network.

DCBCCC



mRNAScoremRNAScoremRNAScore
JUN59JUN33383.63JUN0.069956
ACACA52ACACA23963.01FOS0.069231
FOS51CTNNB118141.12CTNNB10.069133
CDK145FOS16604.96DECR10.068756
DECR142DECR115936.45MAPK30.068542
CTNNB141CDK112244.42ACACA0.068531
MAPK336IL811466.93ACTA10.068425
ACTA132MAPK310861.69ACTA20.068383
ENO331ACTA19482.17CREM0.068351
CCNB131GART   8161.233CDK10.068277
IL829MBP   7587.735AR0.068087
ACTA229AR   6690.792GART0.068024
GART28ENO3   6230.197IL80.068014
EHHADH24DCN   6073.046CCNB10.067804
DCN23RTN4IP1   5927.582ENO30.067784

[i] BC, Betweenness centrality; CC, Closeness centrality; DC, Degree Centrality.

Construction of the lncRNA-mRNA regulatory network and functional prediction of DE-lncRNAs

Following prediction of the target genes of the upregulated and downregulated lncRNAs, lncRNA-mRNA regulatory networks were constructed. For the upregulated lncRNAs, the lncRNA-mRNA regulatory network contained 182 nodes (including 37 lncRNAs and 145 target genes) and 672 interactions. For the downregulated lncRNAs, the lncRNA-mRNA regulatory network contained 140 nodes (including 47 lncRNAs and 93 target genes) and 450 interactions, among which AC004540.4 and RP11-279F6.1 were identified to target JUN. The top 10 nodes (including AC004540.4 and RP11-279F6.1) in the lncRNA-mRNA regulatory networks are summarized in Table III. Subsequent to enrichment analysis, the enriched pathways for the target genes of each upregulated and downregulated lncRNA were compared to identify significant pathways.

Table III.

Top 10 nodes in the lncRNA-mRNA regulatory networks for the upregulated and downregulated lncRNAs.

Table III.

Top 10 nodes in the lncRNA-mRNA regulatory networks for the upregulated and downregulated lncRNAs.

UpregulatedDownregulated


SymbolDegreeSymbolDegree
AC004540.460 XLOC_01410347
RP11-279F6.154 C17orf76-AS145
AQP7P153 RP11-279F6.341
XLOC_00789652 AK02714541
AK02528851 RP11-13L2.434
RP1-60O19.147 RP11-120K9.230
TRHDE-AS146 LOC10050580629
RP11-345M22.242 BC07389726
LOC10042273738 RP11-471J12.121
AK05538629RMST14

[i] lncRNA, long non-coding RNA.

Construction of miRNA-lncRNA-mRNA interaction network

The miRNAs (including miR-409-3p and miR-139, which targeted JUN) targeting the DE-mRNAs involved in the PPI network are summarized in Table IV. Subsequent to prediction of the binding sites between DE-lncRNAs and the miRNAs associated with the miRNA-mRNA pairs, an miRNA-lncRNA interaction network was constructed (involving 26 miRNAs, 111 lncRNAs and 224 interactions). In the miRNA-lncRNA interaction network, miR-409-3p and miR-139 interacted with AC004540.4 and RP11-279F6.1, respectively. The top 10 nodes exhibiting the highest degrees are summarized in Table V. Finally, an miRNA-lncRNA-mRNA interaction network was constructed, which contained 249 nodes, including 36 miRNAs, 95 lncRNAs, 118 mRNAs and 845 interactions (Fig. 3). The top 10 nodes [including miR-20a; solute carrier family 30, member 10, (SLC30A10); and BCL2L11] with the highest degrees in the miRNA-lncRNA-mRNA interaction network are summarized in Table VI. In particular, BCL2L11 was targeted by miR-20a in the interaction network.

Table IV.

miRNAs targeting the differentially expressed mRNAs involved in the protein-protein interaction network.

Table IV.

miRNAs targeting the differentially expressed mRNAs involved in the protein-protein interaction network.

miRNAGene countP-value
hsa_CAGTATT, miR-200B, miR-200C, miR-42919adjP=0.00004
hsa_ACCAAAG, miR-916adjP=0.00220
hsa_AACATTC, miR-409-3P  8adjP=0.00220
hsa_GGCAGCT, miR-2210adjP=0.00220
hsa_ACTGTAG, miR-139  7adjP=0.00240
hsa_ACATTCC, miR-1, miR-20611adjP=0.00240
hsa_AAAGGAT, miR-501  7adjP=0.00240
hsa_TGTTTAC, miR-30A-5P, miR-30C, miR-30D, miR-30B, miR-30E-5P16adjP=0.00240
hsa_GTGCCTT, miR-50619adjP=0.00240
hsa_TACTTGA, miR-26A, miR-26B11adjP=0.00240
hsa_ACTTTAT, miR-142-5P11adjP=0.00240
hsa_GTGCCAT, miR-183  8adjP=0.00360
hsa_TGGTGCT, miR-29A, miR-29B, miR-29C14adjP=0.00670
hsa_ACTGTGA, miR-27A, miR-27B13adjP=0.00680
hsa_GCACTTT, miR-17-5P, miR-20A, miR-106A, miR-106B, miR-20B, miR-519D15adjP=0.00700
hsa_GTGCAAT, miR-25, miR-32, miR-92, miR-363, miR-36710adjP=0.00760
hsa_TCTGATC, miR-383  4adjP=0.00820
hsa_CCTGTGA, miR-513  6adjP=0.00920

[i] hsa, Homo sapiens; miR, microRNA; adjP, adjusted P-value.

Table V.

Top 10 nodes with highest degrees in the miRNA-lncRNA interaction network.

Table V.

Top 10 nodes with highest degrees in the miRNA-lncRNA interaction network.

miRNAlncRNA


SymbolDegreeSymbolDegree
hsa-miR-92a32 C17orf76-AS19
hsa-miR-36721 AC079776.28
hsa-miR-20b21 RP4-669L17.46
hsa-miR-17-5p21 XLOC_0012235
hsa-miR-14518 XLOC_0006385
hsa-miR-2515 RP11-365O16.35
hsa-miR-20a15 RP11-315I20.15
hsa-miR-106a11 RP11-261C10.35
hsa-miR-50110 RP11-249C24.35
hsa-miR-106b  9 BC0059275

[i] hsa, Homo sapiens; miRNA/miR, microRNA; lncRNA, long non-coding RNA.

Table VI.

Top 10 nodes with highest degrees in the miRNA-lncRNA-mRNA interaction network.

Table VI.

Top 10 nodes with highest degrees in the miRNA-lncRNA-mRNA interaction network.

miRNAlncRNAmRNA



SymbolDegreeSymbolDegreeSymbolDegree
hsa-miR-9242AQP7P115FOS25
hsa-miR-20b36 C17orf76-AS114JUN24
hsa-miR-17-5p36 AK055386  8BCL2L1120
hsa-miR-36731 RP11-365O16.3  7ACACA19
hsa-miR-20a30 AC079776.2  7STK3917
hsa-miR-106a26 SLC30A10  6SLC38A215
hsa-miR-2525 RP4-669L17.4  6CDK1615
hsa-miR-106b24 XLOC_014103  5GFPT214
hsa-miR-50621 XLOC_000638  5PTHLH14
hsa-miR-200c20 XLOC_001223  5RPS6KA213

[i] Hsa, Homo sapiens; miRNA/miR, microRNA; lncRNA, long non-coding RNA.

RP11-279F6.1 and AC004540.4 expression

The RT-qPCR results revealed that RP11-279F6.1 and AC004540.4 expression levels were markedly enhanced in the liver tissues from patients with NAFLD compared with the control liver samples (Fig. 4; P<0.01).

Discussion

In the present study, 318 DE-lncRNAs, including 105 upregulated and 213 downregulated lncRNAs, and 609 DE-mRNAs, including 353 upregulated and 256 downregulated mRNAs, were screened in the NAFLD liver tissues compared with normal liver tissues. In the PPI network for DE-mRNAs, JUN, the targeting gene of BCL2L11, was among the top 15 nodes. JUN was targeted by the lncRNAs RP11-279F6.1 and AC004540.4 and miRNAs miR-409-3p and miR-139. Additionally, miR-409-3p and miR-139 were predicted as the DE-mRNAs involved in the PPI network. Additionally, miR-409-3p and miR-139 were regulated by AC004540.4 and RP11-279F6.1, respectively. In the miRNA-lncRNA-mRNA interaction network, miR-20a, SLC30A10 and BCL2L11 were among the top 10 nodes. RP11-279F6.1 and AC004540.4 expression was markedly increased in the NAFLD patient liver tissues compared with the control liver samples.

JUN, also known as AP1, was identified to be increased in a previous study of NAFLD (35). Phosphorylation of the transcriptional activation domain of AP1 is conducted by JNKs to enhance its activity, thereby accelerating the progression and development of NASH (36,37). JUN is considered to be an oncogene in the liver, and its expression is enhanced in response to inflammatory stimuli, promoting liver tumorigenesis (38,39). JUN serves an important role in hepatitis B virus-associated tumorigenesis by promoting the proliferation of hepatocytes and dysplasia progression, indicating that JUN is a useful treatment target for preventing hepatitis-associated tumorigenesis (40). The results from the present study indicated that JUN is involved in the pathogenesis of NAFLD.

In the present study, JUN interacted with BCL2L11 and was targeted by miR-20a in the miRNA-lncRNA-mRNA interaction network. The serum/plasma level of miR-20a has potential value for detecting hepatitis C virus (HCV) infection, and therefore circulating miR-20a may be useful as a predictive marker in liver fibrosis mediated by HCV (41). Apoptosis is a major cause of hepatocyte elimination in NAFLD, and inhibition of the anti-apoptotic protein Bcl-2 and activation of the pro-apoptotic protein p53 promotes inflammation in NAFLD (42). Overexpression of Bcl-2 results in resistance to reperfusion injury in the ischemic liver by suppressing apoptosis and is associated with increased caspase 3 and cytoplasmic cytochrome c and a deficiency of Bcl-extra large (43). By targeting the anti-apoptotic gene Bcl-2, miR-15b and miR-16 regulate tumor necrosis factor-mediated hepatic apoptosis in the process of acute liver failure (44). The data from the present study suggest that miR-20a serves a role in NAFLD by targeting BCL2L11 and affecting the expression of JUN.

JUN was also regulated by the miRNAs of miR-409-3p and miR-139. In hepatoma HuH-7 cells, miR-409-3p decreased the production of fibrinogen by downregulating fibrinogen beta chain precursor expression (45). A previous study suggested that miR-409-3p may be utilized to detect the progression of NAFLD (46). miR-409-3p was also identified as a biomarker for the therapeutics and diagnosis of a number of heart failure-associated diseases and a risk factor of NAFLD (47). Overexpression of miR-139, which was downregulated in hepatocellular carcinoma (HCC) samples, suppresses the progression and metastasis of HCC by downregulating rho-kinase 2, indicating that miR-139 may be used to predict the outcome of HCC (48). In the present study, miR-409-3p and miR-139 interacted with the lncRNAs of AC004540.4 and RP11-279F6.1, respectively. Furthermore, the expression levels of AC004540.4 and RP11-279F6.1 were markedly enhanced in liver tissues from patients with NAFLD compared with the control liver samples. Therefore, we hypothesized that miR-409-3p, miR-139, AC004540.4 and RP11-279F6.1 co-regulated JUN expression in patients with NAFLD.

Although the present study succeeded in identifying specific key miRNAs and lncRNAs in the development of NAFLD, there were also certain limitations. For example, the analyses were based on the microarray dataset GSE72756 downloaded from the GEO database. However, the stages of NAFLD (fatty liver, steatohepatitis or fibrosis/cirrhosis) were not clearly described in the GEO database. Therefore, the degree of NAFLD was not clear. In addition, the sample size was too small, and the predicted regulatory associations were not validated. Future studies will aim to confirm the predicted regulatory associations using cell line experiments.

In conclusion, 318 DE-lncRNAs and 609 DE-mRNAs were identified in NAFLD liver tissues by bioinformatics analysis. Additionally, specific mRNAs (including JUN and BCL2L11) and miRNAs (including miR-20a, miR-409-3p and miR-139) may serve essential roles in the pathogenesis of NAFLD. The lncRNAs AC004540.4 and RP11-279F6.1 were also implicated in the mechanisms of NAFLD.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

HW and XS were responsible for the conception and design of the research and drafting the manuscript. ZT and YL performed the data acquisition. JZ performed the data analysis and interpretation. YS participated in the design of the study and performed the statistical analysis. All authors have read and approved the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Nanjing First Hospital. Informed consent was obtained from all patients.

Patient consent for publication

Informed consent was obtained from all patients.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

lncRNAs

long non-coding RNA

miRNAs

microRNA

DE-lncRNAs

differentially expressed lncRNAs

PPI

protein-protein interaction

NAFLD

non-alcoholic fatty liver disease

NASH

non-alcoholic steatohepatitis

FATP5

fatty acid transport protein 5

FDR

false discovery rate

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

CC

Closeness centrality

DC

Degree Centrality

BC

Betweenness centrality

RT-qPCR

reverse transcription quantitative polymerase chain reaction

BP

biological process

MF

molecular function

CC

cellular component

HCC

hepatocellular carcinoma

References

1 

Shaker M, Tabbaa A, Albeldawi M and Alkhouri N: Liver transplantation for nonalcoholic fatty liver disease: New challenges and new opportunities. World J Gastroenterol. 20:5320–5330. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Rinella ME: Nonalcoholic fatty liver disease: A systematic review. JAMA. 313:22632015. View Article : Google Scholar : PubMed/NCBI

3 

Tolman KG and Dalpiaz AS: Treatment of non-alcoholic fatty liver disease. Ther Clin Risk Manag. 3:1153–1163. 2007.PubMed/NCBI

4 

Clark JM and Diehl AM: Nonalcoholic fatty liver disease: An underrecognized cause of cryptogenic cirrhosis. JAMA. 289:3000–3004. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Omagari K, Kadokawa Y, Masuda J, Egawa I, Sawa T, Hazama H, Ohba K, Isomoto H, Mizuta Y, Hayashida K, et al: Fatty liver in non-alcoholic non-overweight Japanese adults: Incidence and clinical characteristics. J Gastroenterol Hepatol. 17:1098–1105. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Shen L, Fan JG, Shao Y, Zeng MD, Wang JR, Luo GH, Li JQ and Chen SY: Prevalence of nonalcoholic fatty liver among administrative officers in Shanghai: An epidemiological survey. World J Gastroenterol. 9:1106–1110. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Lazo M, Hernaez R, Bonekamp S, Kamel IR, Brancati FL, Guallar E and Clark JM: Non-alcoholic fatty liver disease and mortality among US adults: Prospective cohort study. BMJ. 343:d68912011. View Article : Google Scholar : PubMed/NCBI

8 

Doege H, Grimm D, Falcon A, Tsang B, Storm TA, Xu H, Ortegon AM, Kazantzis M, Kay MA and Stahl A: Silencing of hepatic fatty acid transporter protein 5 in vivo reverses diet-induced non-alcoholic fatty liver disease and improves hyperglycemia. J Biol Chem. 283:22186–22192. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Hoekstra M, Li ZS, Kruijt JK, Van Eck M, Van Berkel TJ and Kuiper J: The expression level of non-alcoholic fatty liver disease-related gene PNPLA3 in hepatocytes is highly influenced by hepatic lipid status. J Hepatol. 52:244–251. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Lin YC, Chang PF, Hu FC, Yang WS, Chang MH and Ni YH: A common variant in the PNPLA3 gene is a risk factor for non-alcoholic fatty liver disease in obese Taiwanese children. J Pediatr. 158:740–744. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Tang Y, Bian Z, Zhao L, Liu Y, Liang S, Wang Q, Han X, Peng Y, Chen X, Shen L, et al: Interleukin-17 exacerbates hepatic steatosis and inflammation in non-alcoholic fatty liver disease. Clin Exp Immunol. 166:281–290. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Yamada H, Suzuki K, Ichino N, Ando Y, Sawada A, Osakabe K, Sugimoto K, Ohashi K, Teradaira R, Inoue T, et al: Associations between circulating microRNAs (miR-21, miR-34a, miR-122 and miR-451) and non-alcoholic fatty liver. Clin Chim Acta. 424:99–103. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Sun C, Huang F, Liu X, Xiao X, Yang M, Hu G, Liu H and Liao L: miR-21 regulates triglyceride and cholesterol metabolism in non-alcoholic fatty liver disease by targeting HMGCR. Int J Mol Med. 35:847–853. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Rui EC, Ferreira DM, Afonso MB, Borralho PM, Machado MV, Cortez-Pinto H and Rodrigues CM: miR-34a/SIRT1/p53 is suppressed by ursodeoxycholic acid in the rat liver and activated by disease severity in human non-alcoholic fatty liver disease. J Hepatol. 58:119–125. 2012.PubMed/NCBI

15 

Sun C, Liu X, Yi Z, Xiao X, Yang M, Hu G, Liu H, Liao L and Huang F: Genome-wide analysis of long noncoding RNA expression profiles in patients with non-alcoholic fatty liver disease. IUBMB Life. 67:847–852. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Smyth GK: Limma: Linear models for microarray data. In: Bioinformatics and Computational Biology Solutions Using R and Bioconductor. Gentleman R, Carey VJ, Huber W, Irizarry RA and Dudoit S: Springer New York. (New York, NY). 397–420. 2005.

17 

Kolde R and Kolde MR: Package ‘pheatmap’. https://cran.r-project.org/web/packages/pheatmap/October 12–2015

18 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Tweedie S, Ashburner M, Falls K, Leyland P, McQuilton P, Marygold S, Millburn G, Osumi-Sutherland D, Schroeder A, Seal R, et al: FlyBase: Enhancing drosophila gene ontology annotations. Nucleic Acids Res 37 (Database Issue). D555–D559. 2009. View Article : Google Scholar

20 

Altermann E and Klaenhammer TR: PathwayVoyager: Pathway mapping using the Kyoto encyclopedia of genes and genomes (KEGG) database. BMC Genomics. 6:602005. View Article : Google Scholar : PubMed/NCBI

21 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43 (Database Issue). D447–D452. 2015. View Article : Google Scholar

22 

Saito R, Smoot ME, Ono K, Ruscheinski J, Wang PL, Lotia S, Pico AR, Bader GD and Ideker T: A travel guide to Cytoscape plugins. Nat Methods. 9:1069–1076. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Tang Y, Li M, Wang J, Pan Y and Wu FX: CytoNCA: A cytoscape plugin for centrality analysis and evaluation of biological networks. Biosystems. 127:67–72. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Du Y, Gao C, Chen X, Hu Y, Sadiq R and Deng Y: A new closeness centrality measure via effective distance in complex networks. Chaos. 25:0331122015. View Article : Google Scholar : PubMed/NCBI

25 

Opsahl T, Agneessens F and Skvoretz J: Node centrality in weighted networks: Generalizing degree and shortest paths. Social Networks. 32:245–251. 2010. View Article : Google Scholar

26 

Cukierski WJ and Foran DJ: Using betweenness centrality to identify manifold Shortcuts. Proc IEEE Int Conf Data Min. 2008:949–958. 2008.PubMed/NCBI

27 

He X and Zhang J: Why do hubs tend to be essential in protein networks? PLoS Genet. 2:e882006. View Article : Google Scholar : PubMed/NCBI

28 

Mukaka MM: Statistics corner: A guide to appropriate use of correlation coefficient in medical research. Malawi Med J. 24:69–71. 2012.PubMed/NCBI

29 

Wang J, Duncan D, Shi Z and Zhang B: WEB-based GEne SeT AnaLysis Toolkit (WebGestalt): Update 2013. Nucleic Acids Res. 41:W77–W83. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM and Haussler D: The human genome browser at UCSC. Genome Res. 12:996–1006. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Kozomara A and Griffiths-Jones S: miRBase: Annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res 42 (Database Issue). D68–D73. 2014. View Article : Google Scholar

32 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Correction: Human MicroRNA targets. PLoS Biol. 3:e2642005. View Article : Google Scholar :

33 

Krüger J and Rehmsmeier M: RNAhybrid: microRNA target prediction easy, fast and flexible. Nucleic Acids Res. 34:W451–W454. 2006. View Article : Google Scholar : PubMed/NCBI

34 

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

35 

Dorn C, Engelmann JC, Saugspier M, Koch A, Hartmann A, Müller M, Spang R, Bosserhoff A and Hellerbrand C: Increased expression of c-Jun in nonalcoholic fatty liver disease. Lab Invest. 94:394–408. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Singh R, Wang YJ, Xiang YQ, Tanaka KE, Gaarde WA and Czaja MJ: Differential effects of JNK1 and JNK2 inhibition on murine steatohepatitis and insulin resistance. Hepatology. 49:87–96. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Karin M and Gallagher E: From JNK to pay dirt: Jun kinases, their biochemistry, physiology and clinical importance. IUBMB Life. 57:283–295. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Min L, Ji Y, Bakiri L, Qiu Z, Cen J, Chen X, Chen L, Scheuch H, Zheng H, Qin L, et al: Liver cancer initiation is controlled by AP-1 through SIRT6-dependent inhibition of survivin. Nat Cell Biol. 15:1203–1211. 2013. View Article : Google Scholar

39 

Machida K, Tsukamoto H, Liu JC, Han YP, Govindarajan S, Lai MM, Akira S and Ou JH: c-Jun mediates hepatitis C virus hepatocarcinogenesis through signal transducer and activator of transcription 3 and nitric oxide-dependent impairment of oxidative DNA repair. Hepatology. 52:480–492. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Trierweiler C, Hockenjos B, Zatloukal K, Thimme R, Blum HE, Wagner EF and Hasselblatt P: The transcription factor c-JUN/AP-1 promotes HBV-related liver tumorigenesis in mice. Cell Death Differ. 23:576–582. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Shrivastava S, Petrone J, Steele R, Lauer GM, Di Bisceglie AM and Ray RB: Up-regulation of circulating miR-20a is correlated with hepatitis C virus-mediated liver disease progression. Hepatology. 58:863–871. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Panasiuk A, Dzieciol J, Panasiuk B and Prokopowicz D: Expression of p53, Bax and Bcl-2 proteins in hepatocytes in non-alcoholic fatty liver disease. World J Gastroenterol. 12:6198–6202. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Selzner M, Rüdiger HA, Selzner N, Thomas DW, Sindram D and Clavien PA: Transgenic mice overexpressing human B cl-2 are resistant to hepatic ischemia and reperfusion. J Hepatol. 36:218–225. 2002. View Article : Google Scholar : PubMed/NCBI

44 

An F, Gong B, Wang H, Yu D, Zhao G, Lin L, Tang W, Yu H, Bao S and Xie Q: miR-15b and miR-16 regulate TNF mediated hepatocyte apoptosis via BCL2 in acute liver failure. Apoptosis. 17:702–716. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Fort A, Borel C, Migliavacca E, Antonarakis SE, Fish RJ and Neerman-Arbez M: Regulation of fibrinogen production by microRNAs. Blood. 116:2608–2615. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Tryndyak VP, Marrone AK, Latendresse JR, Muskhelishvili L, Beland FA and Pogribny IP: MicroRNA changes, activation of progenitor cells and severity of liver injury in mice induced by choline and folate deficiency. J Nutr Biochem. 28:83–90. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Yang Y, Yu T, Jiang S, Zhang Y, Li M, Tang N, Ponnusamy M, Wang JX and Li PF: miRNAs as potential therapeutic targets and diagnostic biomarkers for cardiovascular disease with a particular focus on WO2010091204. Expert Opin Ther Pat. 27:1021–1029. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Wong CC, Wong CM, Tung EK, Au SL, Lee JM, Poon RT, Man K and Ng IO: The microRNA miR-139 suppresses metastasis and progression of hepatocellular carcinoma by down-regulating Rho-kinase 2. Gastroenterology. 140:322–331. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2019
Volume 19 Issue 4

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
Wu H, Song X, Ling Y, Zhou J, Tao Z and Shen Y: Comprehensive bioinformatics analysis of critical lncRNAs, mRNAs and miRNAs in non‑alcoholic fatty liver disease. Mol Med Rep 19: 2649-2659, 2019
APA
Wu, H., Song, X., Ling, Y., Zhou, J., Tao, Z., & Shen, Y. (2019). Comprehensive bioinformatics analysis of critical lncRNAs, mRNAs and miRNAs in non‑alcoholic fatty liver disease. Molecular Medicine Reports, 19, 2649-2659. https://doi.org/10.3892/mmr.2019.9931
MLA
Wu, H., Song, X., Ling, Y., Zhou, J., Tao, Z., Shen, Y."Comprehensive bioinformatics analysis of critical lncRNAs, mRNAs and miRNAs in non‑alcoholic fatty liver disease". Molecular Medicine Reports 19.4 (2019): 2649-2659.
Chicago
Wu, H., Song, X., Ling, Y., Zhou, J., Tao, Z., Shen, Y."Comprehensive bioinformatics analysis of critical lncRNAs, mRNAs and miRNAs in non‑alcoholic fatty liver disease". Molecular Medicine Reports 19, no. 4 (2019): 2649-2659. https://doi.org/10.3892/mmr.2019.9931