Open Access

Integration of high‑throughput data of microRNA and mRNA expression profiles reveals novel insights into the mechanism of liver fibrosis

  • Authors:
    • Yitong Zhang
    • Jing Liu
    • Yanyun Ma
    • Jingjie Wang
    • Jie Zhu
    • Jie Liu
    • Jun Zhang
  • View Affiliations

  • Published online on: November 9, 2018     https://doi.org/10.3892/mmr.2018.9641
  • Pages: 115-124
  • Copyright: © Zhang 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

Numerous studies have revealed that microRNAs (miRNAs) are functional non‑coding RNAs that serve roles in a variety of biological processes. However, the expression patterns and regulatory networks, as well as the miRNAs involved in liver fibrosis remain to be elucidated. In the present study, a mouse model of liver fibrosis was constructed by CCl4 intraperitoneal injection and the total RNAs were extracted from the liver of the mice. The total RNAs were then sequenced on an Illumina HiSeq 2000 platform and an integrated analysis of miRNA and mRNA expression profiles in CCl4‑induced liver fibrosis was performed. Compared with normal liver samples, 56 and 15 miRNAs were found to be upregulated and downregulated in fibrotic livers, respectively. To predict the potential functions of these miRNAs, bioinformatics analysis, including Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis, was used to assess target mRNAs. The results indicated that the mitogen‑activated protein kinase, phosphoinositide 3 kinase/protein kinase B and focal adhesion signaling pathways were the most significantly enriched. In addition, a regulatory network containing five dysregulated miRNAs and 22 target mRNAs was constructed based on their inverse correlation. Furthermore, the five dysregulated miRNAs were significantly upregulated and the expression of RELB, RAP1A, PPP3CB, MAP2K4, ARRB1, MAP3K4, FGF1 and PRKCB in the network was significantly decreased in LX‑2 cells following TGF‑β1 treatment which suggested that they were associated with the activation of human hepatic stellate cells. The miRNA‑mRNA regulatory network produced in the present study may provide novel insights into the role of miRNAs in liver fibrosis.

Introduction

Hepatic fibrosis is an inevitable process that occurs in the progression of various chronic liver diseases, ultimately leading to cirrhosis. It is a reversible pathological change caused by acute/chronic liver injury (1). Liver fibrosis manifests as the excessive deposition of extracellular matrix (ECM) in the liver. Long-term aggravation of liver fibrosis results in diffuse liver damage, which may lead to the development of cirrhosis and liver cancer (1). The activation and proliferation of hepatic stellate cells (HSCs) is critically involved in the process of liver fibrosis (24). It is therefore of great importance to identify a reliable, non-invasive and convenient method for the early diagnosis of liver fibrosis.

MicroRNAs (miRNAs/miRs) are a class of evolutionarily conserved small (18–24 nucleotides) single-stranded non-coding RNAs that mediate post-transcriptional gene suppression, via the degradation of target mRNAs or the suppression of mRNA translation following binding (5,6). Increasing evidence suggests that dysregulated miRNAs serve a crucial role in a number of diseases, including hepatocellular carcinoma and liver fibrosis (7,8). For example, it was reported that the miRNA-15a/plasminogen activator inhibitor 2 axis promotes the migration of cholangiocarcinoma cells (9). miR-122 and other miRNAs may have potential as circulating biomarkers in drug-induced liver injury (10). miRNA-351 directly targets the vitamin D receptor to promote schistosomiasis-induced hepatic fibrosis (11). However, to the best of our knowledge, there have been no studies focusing on the miRNA-mRNA network involved in liver fibrosis to date.

In the current study, the expression profiles of miRNAs and mRNAs in liver fibrosis were investigated and compared with normal liver samples. The identification of novel differentially expressed miRNAs may provide novel insights, allowing for the early diagnosis and treatment of liver fibrosis. A total of 71 differentially expressed miRNAs (including 56 upregulated and 15 downregulated miRNAs) in were identified fibrotic tissues compared with normal liver tissues. Integrated analysis with human liver cirrhosis data from Gene Expression Omnibus (GEO) datasets revealed 5 miRNAs that were significantly increased in both human and mouse fibrotic liver tissues. A functional miRNA-mRNA network was constructed based on their inverse correlation. Target mRNAs identified in this network were further confirmed in activated hepatic stellate cells (HSCs). The results of the present study may provide novel insights to increase our understanding of the underlying mechanism involved in liver fibrosis.

Materials and methods

Liver fibrosis model and RNA isolation

A mouse model of liver fibrosis was constructed and RNA was isolated as previously reported (12). In brief, 6-week-old male C57BL/6 mice (weight, ~20 g) were purchased from Shanghai Laboratory Animal Center (Shanghai, China) and maintained under a 12 h light/dark cycle with 40–60% humidity, at 22–25°C, with free access to food and water. Following acclimatization for one week, the 12 mice were randomly divided into control and treatment groups (n=6/each group). CCl4 (0.5 µl/g body weight; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) diluted nine times in corn oil (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was administered to mice via intraperitoneal injection by two times per week, for 8 weeks. The control group was injected with an equivalent volume of corn oil. After 8 weeks, mice were anaesthetized by intraperitoneal injection of 0.8% pentobarbital (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). and sacrificed and liver tissues were harvested. Total RNA was isolated from four fibrotic and four normal liver tissues using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's instructions. The study was approved by the Ethical Committee of Fudan University and all experiments were performed according to the approved guidelines and regulations.

Histopathology analyses and immunohistochemistry

Liver specimens were fixed in 4% formaldehyde (Sigma-Aldrich; Merck KGaA) at room temperature for 12 h, dehydrated, embedded in paraffin at room temperature for 1 h and sectioned at 3 um. The sections were processed for hematoxylin and eosin (H&E) and Masson's trichrome (both Sigma-Aldrich; Merck KGaA) staining for assessment of the degree of liver fibrosis. For the H&E staining, the tissue section was incubated at 65°C for 1 h for dewaxing. Samples were stained with hematoxylin for 1 min, followed by eosin staining for 10 sec at room temperature, and, then the samples were dried at room temperature and sealed. The middle part of the visual field was examined by light microscopy. Masson Trichrome staining was performed following the protocol of trichrome stain (Masson) kit from Sigma-Aldrich. Deparaffinized the slides to deionized water, then mordanted in preheated Bouin's Solution at 56°C for 15 min. Cooled the slides in tap water (18–26 °C) and washed in running tap water to remove yellow color from sections. Put the slides in Working Weigert's Iron Haematoxylin Solution for 5 min, Biebrich Scarlet-Acid Fucshin for 5 min, working Phosphotungstic/Phosphomolybdic Acid Solution for 5 min, Aniline Blue Solution for 5 min, 1% acetic acid for 2 min both at room temperature. Finally, rinsed slides, dehydrated through alcohol and cleared in xylene. A second set of tissue sections (0.5×0.5 cm) were prepared for immunohistochemical assessment of the tissues. Briefly, endogenous peroxidase activity was blocked by immersion of deparaffinized sections in 3% H2O2 in methanol for 30 min at room temperature. Antigen retrieval was performed by steaming slides in 0.01 M citrate buffer (pH 6.0) and heated them until the temperature reached 95–100°C for 30 min. The samples were incubated for 30 min at room temperature in 5% normal blocking serum, and incubated with a 1:100 dilution of polyclonal rabbit anti-α-SMA (1:200, cat. no. ab5694, Abcam, Cambridge, UK) overnight at 4°C. The slides were then incubated with HRP-conjugated goat anti-rabbit secondary antibody (1:2,000, ab7090; Abcam) for 60 min at room temperature. Between each incubation, sections were washed three times with PBS. Sections were developed with 3,3-diaminobenzidine tetrahydrochloride and hydrogen peroxide and subsequently counterstained with hematoxylin. The sections were imaged using a Nikon Eclipse 80i microscope (Nikon Corporation, Tokyo, Japan). Hematoxylin and eosin, α-smooth muscle actin (SMA) and Masson staining demonstrated that liver fibrosis was successfully established, which was also verified in our previous study (12).

miRNA sequencing

Total RNA was used to prepare miRNA libraries and the purified libraries were sequenced on an Illumina HiSeq 2000 platform (Illumina, Inc., San Diego, CA, USA). In brief, the raw data were processed with R software (version 3.3.2; www.r-project.org) to ensure its quality. Clean data were mapped to the Ensemble database (GRCh37) (13) and compared with miRBase (release 21; www.mirbase.org) to identify mature miRNAs. The count and reads per million total read values of the miRNAs were collected for each sample. Individual miRNAs were analyzed to identify significant differences using the DESeq2 package (version 1.4.0) (14) in R. The parameters for differentially expressed miRNAs were set with a false discovery rate of <0.05 and |log2 fold change (FC)|>=1. The differentially expressed miRNAs are shown by heatmap and volcano plot using R software The miRNA expression profiles (GSE49012 and GSE40744) in human liver cirrhosis tissues were downloaded from publicly available Gene Expression Omnibus (GEO) datasets (www.ncbi.nlm.nih.gov/geo) (15,16).

miRNA target prediction

miRWalk (version 2.0) (17) collects 13 predicted data sets from existing miRNA target databases to predict all possible miRNA-mRNA interactions. Putative interactions between the sequenced miRNAs and mRNAs were evaluated using miRWalk. The search was restricted to miRNAs with a minimum seed length of seven nucleotides and binding sites in the 3′untranslated region (UTR) of target genes. P<0.05 was considered to indicate a statistically significant difference.

Functional analysis

To explore the functional roles of target genes, the Database for Annotation, Visualization and Integrated Discovery (version 6.8; www.david.ncifcrf.gov) software (18,19) was used, which integrates the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases to analyze biological functions. Fisher's exact test was performed to evaluate the enrichment values of GO terms and KEGG pathways. P<0.05 was considered to indicate a statistically significant difference. The miRNA-mRNA interaction regulatory network was constructed using Cytoscape software (20) (version 3.3.0; www.cytoscape.org).

Activation of LX-2 cells

LX-2 HSC cells were cultured in DMEM (Invitrogen; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (FBS, Invitrogen; Thermo Fisher Scientific, Inc.) in a humidified atmosphere containing 5% CO2 at 37°C for 24 h. LX-2 cells were starved for 12 h in serum-free DMEM, following which they were treated with 10 ng/ml TGF-β (R&D Systems China Co., Ltd., Shanghai, China). After 24 h of TGF-β treatment, the cells were harvested for RNA isolation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR).

RT-qPCR

SYBR Green-based qPCR (Thermo Fisher Scientific, Inc.) was used to measure mRNA expression. Total RNA (2 µg) was reverse transcribed into cDNA using the reverse transcription kit (Takara Biotechnology Co., Ltd. Dalian, China). The protocol for reverse transcription was as follows: 25°C for 10 min; 37°C for 120 min and 85°C for 5 min. The thermocycling conditions for RT-qPCR were as follows: Initial denaturation at 95°C for 10 min; 40 cycles of denaturation at 95°C for 30 sec, annealing at 55°C for 30 sec and extension at 72°C for 30 sec; followed by melting curve analysis. All experiments were performed in triplicate. The comparative ∆Cq method (21) was used to analyze the data, with GAPDH as an endogenous reference gene. The RT-PCR primers are listed in Table I.

Table I.

Primers used for RT-qPCR.

Table I.

Primers used for RT-qPCR.

Gene (human)Forward primer (5′to 3′)Reverse primer (5′to 3′)
RELB CAGCCTCGTGGGGAAAGAC GCCCAGGTTGTTAAAACTGTGC
  RAP1A CGTGAGTACAAGCTAGTGGTCC CCAGGATTTCGAGCATACACTG
  PPP3CB CCCCAACACATCGCTTGACAT GGCAGCACCCTCATTGATAATTC
  ARRB1 AAAGGGACCCGAGTGTTCAAG CGTCACATAGACTCTCCGCT
  MAP2K4 TGCAGGGTAAACGCAAAGCA CTCCTGTAGGATTGGGATTCAGA
  FGF1 CTCCCGAAGGATTAAACGACG GTCAGTGCTGCCTGAATGCT
  MAP3K4 CTCGACAGATGAAACGCATGT CCAGTGTCTTTATGTGGAGGC
  PPKCB AGCCCCACGTTTTGTGACC GCTGGGAACATTCATCACGC
  GAPDH ACAACTTTGGTATCGTGGAAGG GCCATCACGCCACAGTTTC
Gene (mouse)
  Relb CACCGGGTACACCCACATAG ATGCCCAGGTTGTTAAAGCTG
  Rap1a ATGCGTGAGTACAAGCTAGTAGT AATCTACCTCGACTTGCTTTCTG
  Ppp3cb AAAGCGTGCTGACACTCAAG TGGAGAGAATCCTCGTATTGCT
  Arrb1 AGGCAAGCCCCAATGGAAAG AGTGTCACGTAGACTCGCCTT
  Map2k4 AATCGACAGCACGGTTTACTC GCAGTGAAATCCCAGTGTTGTT
  Fgf1 GGGGAGATCACAACCTTCGC GTCCCTTGTCCCATCCACG
  Map3k4 GAGTCGGCTCGCAAAAGTATG GTGAGGTGCCGTAGAGAGTC
  Ppkcb ATGAGTTCGTCACGTTCTCCT CCATACAGCAGCGATCCACAG
  Gapdh AGGTCGGTGTGAACGGATTTG GGGGTCGTTGATGGCAACA
Western blotting

Protein samples were extracted from cells using radioimmunoprecipitation lysis buffer (Beyotime Biotechnology, Inc., Haimen, China). Total protein concentration was quantified by Bicinchoninic acid Protein Assay kit (Beyotime Biotechnology). Equal amounts of protein (30 µg/lane) were separated on 8–12% polyacrylamide gels and transferred onto polyvinylidene difluoride membranes. Subsequently, the membranes were blocked with 5% fat-free dry milk at room temperature for 1 h. Membranes were incubated with mouse anti-α-SMA (cat. no. A5228, 1:1,000; Sigma-Aldrich; Merck KGaA), anti-collagen α-1(I) chain (COL1α1; cat. no. ABT123, 1:1,000, EMD Millipore, Billerica, MA, USA) or anti-GAPDH (cat. no. ab8245, 1:4,000; Abcam, Cambridge, UK) antibodies in Tris buffered saline with 0.1% Tween-20 (TBST) overnight on a shaker at 4°C. Membranes were subsequently washed three times in TBST and incubated with horseradish peroxidase (HRP)-conjugated rabbit anti-mouse IgG secondary antibodies (1:5,000, cat. no. ab6728, Abcam) at room temperature for 1 h. Immunoblots were visualized using Enhanced Chemiluminescent Plus western blotting substrate (Thermo Fisher Scientific, Inc.).

Statistical analysis

Student's t-test was performed to compare two variables using SPSS v20 software (IBM Corp., Armonk, NY, USA). All data are expressed as the mean ± standard deviation. FC≥2 and P<0.05 were considered to indicate a statistically significant difference. All experiments were repeated in triplicate.

Results

Identification of differentially expressed miRNAs in liver fibrosis

To identify miRNAs that may be significant in liver fibrogenesis, miRNA sequencing was performed to compare their expression between fibrotic liver and normal liver tissues. As shown in Fig. 1, 71 miRNAs that were significantly differentially expressed between fibrotic liver and normal liver tissues were identified. Among these, 56 were upregulated and 15 were downregulated in fibrotic samples. A heatmap and volcano plot were produced to show the differentially expressed miRNAs using R software, of which the top 20 dysregulated miRNAs were summarized and listed in Table II. To identify conserved miRNAs in human clinical samples and animal experiments, two miRNA profiles (GSE49012 and GSE40744) were downloaded from publicly available GEO datasets (www.ncbi.nlm.nih.gov/geo). The data suggested that five miRNAs (miR-212-3p, miR-146b-5p, let-7i-5p, miR-143-5p and miR-199a-3p) were also upregulated in human liver fibrotic samples, which was consistent with the results of the present study (Fig. 1C). RT-qPCR was performed to verify the expression of these five upregulated miRNAs in CCl4-induced fibrotic and control normal liver tissues. It was confirmed that miR-212-3p, miR-146b-5p, let-7i-5p, miR-143-5p and miR-199a-3p were significantly increased in CCl4-treated liver tissues compared with control samples (Fig. 1D).

Table II.

Dysregulated microRNAs in CCl4-induced liver fibrosis (>2-fold; P<0.05).

Table II.

Dysregulated microRNAs in CCl4-induced liver fibrosis (>2-fold; P<0.05).

A, Upregulated

CCl4/Corn Oil CCl4/Corn Oil


Probe IDLog2 changeFold-changeP-value
mmu-miR-212-3p5.0433.00.04778
mmu-miR-3081-3p4.6425.00.03257
mmu-miR-19834.3220.00.02042
mmu-miR-411-3p4.0917.00.03905
mmu-miR-147-5p3.9115.00.03943
mmu-miR-543-3p3.8114.00.02985
mmu-miR-582-3p3.7713.60.00001
mmu-miR-708-3p3.7213.20.00017
mmu-miR-134-5p3.7213.20.00078
mmu-miR-376b-3p3.7013.00.00202

B, Downregulated

CCl4/Corn Oil CCl4/Corn Oil


Probe IDLog2 change Fold-changeP-value

mmu-miR-193a-5p−10.500.02130
mmu-miR-26b-3p−10.500.04648
mmu-miR-137-3p−1.120.460.04818
mmu-miR-871-3−1.190.440.00943
mmu-miR-1948-5p−1.230.430.01038
mmu-miR-365-3p−1.300.410.00174
mmu-miR-192-3p−1.350.390.01571
mmu-miR-741-3p−1.450.370.00290
mmu-miR-6390−1.600.330.00052
mmu-miR-122−1.690.310.00252
GO analysis and pathway analysis

Bioinformatics analyses were used to analyze miRNA function based on their target mRNAs. GO analysis was performed to evaluate mRNA enrichments in terms of biological process, cellular component and molecular function. The top 10 enriched GO terms were involved in the regulation of cellular and metabolic processes (Fig. 2A). KEGG analysis revealed the top 10 pathways, which included the mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt), focal adhesion and Wnt signaling pathways. All of these pathways are associated with HSC activation and extracellular matrix ECM remodeling (2224). These results suggested that some differentially expressed miRNAs may be involved in the progression of liver fibrosis (Fig. 2B).

Construction of the miRNA-mRNA interaction network

To further investigate the association between distinct miRNAs and target mRNAs in fibrotic liver tissues, the mRNA sequencing profile of the CCl4-induced liver fibrosis mouse model was extracted from our previous study (12). A miRNA-mRNA regulatory network associated with liver fibrosis was constructed based on the inverse expression relationships between the five miRNAs upregulated in both mouse and human fibrotic liver tissues and mRNAs significantly enriched in the MAPK, PI3K-Akt, focal adhesion and Wnt signaling pathways. As shown in Fig. 3A, the network contained five miRNAs, 22 mRNAs and 23 miRNA-mRNA connections. Target mRNAs in the network, such as RELB proto-oncogene (RELB), RAS-related protein 1a (RAP1A), protein phosphatase 3 catalytic subunit β (PPP3CB), arrestin β 1 (ARRB1), fibroblast growth factor 1 (FGF1), protein kinase C β (PRKCB) and members of the MAPK family (MAP3K4, MAPK8, MAPKAPK5, MAP2K5, MAP2K4, MAP3K1), were summarized in Table III. The RT-qPCR results revealed that the expression of RELB, RAP1A, PPP3CB, MAP2K4, ARRB1, MAP3K4, FGF1 and PRKCB was decreased in CCl4-induced fibrotic liver tissues (Fig. 3B). The integrated analysis revealed their regulatory interactions in liver fibrosis.

Table III.

Putative target genes involved in the microRNA-mRNA network.

Table III.

Putative target genes involved in the microRNA-mRNA network.

GeneOfficial full nameReference sequenceTargeting miRNA
ARRB1Arrestin β 1NM_178220.3mmu-miR-212-3p
mmu-miR-143-5p
NLKNemo like kinaseNM_008702.3 mmu-miR-146b-5p
PPP3CBProtein phosphatase 3 catalytic subunit βNM_008914.3 mmu-miR-146b-5p
MAPK8Mitogen-activated protein kinase 8NM_001310454.1 mmu-miR-146b-5p
MAP3K4Mitogen-activated protein kinase kinase kinase 4NM_011948.2 mmu-miR-146b-5p
SOS1SOS Ras/Rac guanine nucleotide exchange factor 1NM_009231.2 mmu-miR-146b-5p
FGF1Fibroblast growth factor 1NM_001033789.2 mmu-miR-146b-5p
TRAF6TNF receptor-associated factor 6NM_009424.3 mmu-miR-146b-5p
RAP1ARAS-related protein 1aNM_145541.5 mmu-miR-146b-5p
MAPKAPK5MAP kinase-activated protein kinase 5NM_010765.2 mmu-miR-146b-5p
PPP5Protein phosphatase 5, catalytic subunitNM_011155.2 mmu-miR-146b-5p
CASP3Caspase 3NM_001284409.1 mmu-miR-146b-5p
MAP2Kmitogen-activated protein kinase kinase 5NM_011840.2 mmu-miR-146b-5p
MAP2K4Mitogen-activated protein kinase kinase 4NM_001316368.1mmu-let-7i-5p
DDITDNA-damage inducible transcript 3NM_001003913.2 mmu-miR-199a-3p
MAXMax proteinNM_008558.2 mmu-miR-199a-3p
NR4A1Nuclear receptor subfamily 4NM_010444.2 mmu-miR-199a-3p
SRFSerum response factorNM_020493.2 mmu-miR-199a-3p
IL1BInterleukin 1 βNM_008361.4 mmu-miR-199a-3p
PRKCBProtein kinase C βNM_008855.2 mmu-miR-199a-3p
RELB Reticuloendotheliosis viral (v-rel) oncogene related BNM_009046.2 mmu-miR-199a-3p
MAP3K1Mitogen-activated protein kinase kinase kinase 1NM_011945.2mmu-miR-143-5p
Target mRNAs in the network are associated with HSC activation

HSCs have an essential role in liver fibrosis, in which they undergo activation or transdifferentiation from quiescent to myofibroblast-like cells as a result of injury (25). LX-2 is a widely used human HSC line that can be activated by TGF-β1. To further target mRNAs in HSC activation, the expression of mRNAs in LX-2 cells was examined after 24 h of TGF-β1 (10 ng/ml) stimulation. Following stimulation, the expression of COL1α1 and α-SMA was significantly upregulated (Fig. 4A), suggesting that LX-2 cells were activated. In addition, miR-212-3p, miR-146b-5p, let-7i-5p, miR-143-5p and miR-199a-3p expression was significantly upregulated in LX-2 cells following TGF-β1 treatment (Fig. 4B). Of the 22 mRNAs assessed, the expression of RELB, RAP1A, PPP3CB, MAP2K4, ARRB1, MAP3K4, FGF1 and PRKCB was also significantly decreased (Fig. 4C). The expression pattern of these mRNAs was inverse to that of their regulatory miRNAs (miR-199a-3p for RELB and PRKCB; miR-146b-5p for RAP1A, FGF1 and PPP3CB; miR-143a-3p for MAP3K4; and let-7i-5p for MAP2K4) in the activated HSCs. These results suggested that these miRNAs have an inhibitory effect on their target genes and may serve important roles in liver fibrosis.

Discussion

An increasing number of studies have reported that miRNAs serve a role in regulating the progression and metastasis of various cancers (2630). However, the role of miRNAs and their specific mechanism of action in liver fibrogenesis remain to be elucidated (31). In the present study, miRNA sequencing was performed in CCl4-induced liver fibrosis and integrated with miRNA array data from clinical liver fibrosis samples to compare miRNA expression profiles between fibrotic and normal liver tissues. It was demonstrated that five miRNAs (miR-212-3p, miR-146b-5p, let-7i-5p, miR-199a-3p and miR-143-5p) were significantly upregulated in CCl4-treated livers and human liver cirrhosis samples compared with normal liver tissues. Furthermore, GO and KEGG pathway enrichment analysis was performed for miRNA target genes to identify the signaling pathways involved in hepatic fibrosis. GO enrichment analysis indicated that the majority of the miRNA target genes were involved in cellular metabolic processes closely associated with the onset and development of hepatic fibrosis

The results of KEGG pathway enrichment analysis indicated a total of 159 signaling pathways that may be essential for the development of liver fibrosis. Several of these pathways were already known to be involved in the progression of liver fibrosis, such as the MAPK, focal adhesion, Wnt, p53, mTOR and PI3K-Akt signaling pathways (2224). Various metabolic pathways were also identified in the present study's analysis such as glycan metabolism, cysteine and methionine metabolism and tyrosine metabolism pathway.

HSCs are an important cell type in the process of liver fibrosis, as they can be activated by a large number of cytokines secreted by damaged hepatocytes in chronic liver injury. Activated HSCs promote the deposition of ECM, which in turn leads to the development of liver fibrosis. (32). Therefore, several molecules involved in the activation of HSCs may have potential as therapeutic targets in liver fibrosis (3335). Previous studies have demonstrated that MAPK signaling plays a key role in HSC activation and liver fibrogenesis: MAPK signaling promotes the activation and proliferation of HSCs, increases collagen mRNA stability and leads to the accumulation of ECM (36,37). These findings suggest that dysregulated miRNAs that affect MAPK signaling may affect HSC activation in hepatic fibrosis. In the current study, MAPK-related genes, including RELB, RAP1A, PPP3CB, ARRB1, MAP2K4, FGF1, MAP3K4 and PRKCB, were dysregulated in TGF-β-induced activation of HSCs, suggesting that they may be essential for HSC activation.

A number of studies have reported that miRNAs are involved in the regulation of liver fibrosis (3840). let-7i, miR-342-3p, miR-188-5p and miR-34a are upregulated in CCl4-treated fibrotic livers compared with control samples, while miR-202, miR-378b and miR-378d are downregulated (41). It was previously reported that a module containing five downregulated miRNAs (miR-130b-3p, miR-148a-3p, miR-345-5p, miR-378a-3p and miR-422a) was associated with HSC activation (42). The miR-199 family is closely associated with the progression of liver fibrosis in humans and mice (43). In our previous study, it was demonstrated that the miR-212 promotes HSC activation by targeting Smad family member 7 (44). Similarly, the results of the present study revealed that miR-342-3p, let-7i, miR-212 and miR-199 were significantly upregulated in cirrhotic liver tissues, whereas miR-378a-3p was significantly downregulated. These miRNAs may therefore be promising biomarkers for the early diagnosis of liver cirrhosis.

In conclusion, the miRNA expression profile generated in the present study revealed that the expression of several miRNAs (miR-212-3p, miR-146b-5p, let-7i-5p, miR-143-5p, miR-199a-3p) was associated with liver fibrosis. A miRNA-mRNA network was identified, comprised of 5 miRNAs and 22 mRNAs that were associated with liver fibrosis. This study may provide novel insights and potential biomarkers for the early diagnosis and treatment of liver fibrosis. Future studies should focus on the biological functions of these miRNAs.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (grant nos. 81420108005 and 81300327) and the Ministry of Science and Technology of China (grant no. 2013CB945401).

Availability of data and materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

YZ and JinL performed the cell experiments, analyzed the data and wrote the paper; YM, JW and JieZ conceived the mouse experimental design and performed the animal experiments. JieL and JunZ designed the study and revised the paper.

Ethics approval and consent to participate

The animal study was approved by the Animal Care and Use Committee at the Fudan University. Animals were anesthetized and sacrificed using acceptable methods.

Patient consent for publication

Not applicable.

Competing interests

The authors declare they have no competing interests.

References

1 

Bataller R and Brenner DA: Liver fibrosis. J Clin Invest. 115:209–218. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Friedman SL: Liver fibrosis-from bench to bedside. J Hepatol. 1 Suppl 38:S38–S53. 2003. View Article : Google Scholar

3 

Moreira RK: Hepatic stellate cells and liver fibrosis. Arch Pathol Lab Med. 131:1728–1734. 2007.PubMed/NCBI

4 

Ellis EL and Mann DA: Clinical evidence for the regression of liver fibrosis. J Hepatol. 56:1171–1180. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Chen W and Qin C: General hallmarks of microRNAs in brain evolution and development. RNA Biol. 12:701–708. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP and Bartel DP: MicroRNA targeting specificity in mammals: Determinants beyond seed pairing. Mol Cell. 27:91–105. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Wang XW, Heegaard NH and Orum H: MicroRNAs in liver disease. Gastroenterology. 142:1431–1443. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Szabo G and Bala S: MicroRNAs in liver disease. Nat Rev Gastroenterol Hepatol. 10:542–552. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Utaijaratrasmi P, Vaeteewoottacharn K, Tsunematsu T, Jamjantra P, Wongkham S, Pairojkul C, Khuntikeo N, Ishimaru N, Sirivatanauksorn Y, Pongpaibul A, et al: The microRNA-15a-PAI-2 axis in cholangiocarcinoma-associated fibroblasts promotes migration of cancer cells. Mol Cancer. 17:102018. View Article : Google Scholar : PubMed/NCBI

10 

Howell LS, Ireland L, Park BK and Goldring CE: MiR-122 and other microRNAs as potential circulating biomarkers of drug-induced liver injury. Expert Rev Mol Diagn. 18:47–54. 2018. View Article : Google Scholar : PubMed/NCBI

11 

He X, Sun Y, Lei N, Fan X, Zhang C, Wang Y, Zheng K, Zhang D and Pan W: MicroRNA-351 promotes schistosomiasis-induced hepatic fibrosis by targeting the vitamin D receptor. Proc Natl Acad Sci USA. 115:180–185. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Zhang Y, Liu J, Ma Y, Wang J, Zhu J, Liu J and Zhang J: Integrated profiling of long non-coding RNAs and mRNAs identifies novel regulators associated with liver fibrosis. Pathol Res Pract. 214:1794–1803. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Aken BL, Achuthan P, Akanni W, Amode MR, Bernsdorff F, Bhai J, Billis K, Carvalho-Silva D, Cummins C, Clapham P, et al: Ensembl 2017. Nucleic Acids Res. 45:D635–d642. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Love MI, Huber W and Anders S: Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15:5502014. View Article : Google Scholar : PubMed/NCBI

15 

Vuppalanchi R, Liang T, Goswami CP, Nalamasu R, Li L, Jones D, Wei R, Liu W, Sarasani V, Janga SC and Chalasani N: Relationship between differential hepatic microRNA expression and decreased hepatic cytochrome P450 3A activity in cirrhosis. Plos One. 8:e744712013. View Article : Google Scholar : PubMed/NCBI

16 

Diaz G, Melis M, Tice A, Kleiner DE, Mishra L, Zamboni F and Farci P: Identification of microRNAs specifically expressed in hepatitis C virus-associated hepatocellular carcinoma. Int J Cancer. 133:816–824. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Dweep H and Gretz N: miRWalk2.0: A comprehensive atlas of microRNA-target interactions. Nat Methods. 12:6972015. View Article : Google Scholar : PubMed/NCBI

18 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Huang da W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Britton RS and Bacon BR: Intracellular signaling pathways in stellate cell activation. Alcohol Clin Exp Res. 23:922–925. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Zhao XK, Yu L, Cheng ML, Che P, Lu YY, Zhang Q, Mu M, Li H, Zhu LL, Zhu JJ, et al: Focal adhesion kinase regulates hepatic stellate cell activation and liver fibrosis. Sci Rep. 7:40322017. View Article : Google Scholar : PubMed/NCBI

24 

Wang Y, Ma J, Chen L, Xie XL and Jiang H: Inhibition of focal adhesion kinase on hepatic stellate-cell adhesion and migration. Am J Med Sci. 353:41–48. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Tsuchida T and Friedman SL: Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 14:397–411. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Gantier MP, McCoy CE, Rusinova I, Saulep D, Wang D, Xu D, Irving AT, Behlke MA, Hertzog PJ, Mackay F and Williams BR: Analysis of microRNA turnover in mammalian cells following Dicer1 ablation. Nucleic Acids Res. 39:5692–5703. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, Galas DJ and Wang K: The microRNA spectrum in 12 body fluids. Clin Chem. 56:1733–1741. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Lee HM, Nguyen DT and Lu LF: Progress and challenge of microRNA research in immunity. Front Genet. 5:1782014. View Article : Google Scholar : PubMed/NCBI

29 

Shenoy A and Blelloch RH: Regulation of microRNA function in somatic stem cell proliferation and differentiation. Nat Rev Mol Cell Biol. 15:565–576. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Anthony PP, Ishak KG, Nayak NC, Poulsen HE, Scheuer PJ and Sobin LH: The morphology of cirrhosis. Recommendations on definition, nomenclature, and classification by a working group sponsored by the World Health Organization. J Clin Pathol. 31:395–414. 1978. View Article : Google Scholar : PubMed/NCBI

32 

Zhang CY, Yuan WG, He P, Lei JH and Wang CX: Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J Gastroenterol. 22:10512–10522. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Li D, He L, Guo H, Chen H and Shan H: Targeting activated hepatic stellate cells (aHSCs) for liver fibrosis imaging. EJNMMI Res. 5:712015. View Article : Google Scholar : PubMed/NCBI

34 

Trautwein C, Friedman SL, Schuppan D and Pinzani M: Hepatic fibrosis: Concept to treatment. J Hepatol. 62 Suppl 1:S15–S24. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Mallat A and Lotersztajn S: Reversion of hepatic stellate cell to a quiescent phenotype: From myth to reality? J Hepatol. 59:383–386. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Hattori S, Dhar DK, Hara N, Tonomoto Y, Onoda T, Ono T, Yamanoi A, Tachibana M, Tsuchiya M and Nagasue N: FR-167653, a selective p38 MAPK inhibitor, exerts salutary effect on liver cirrhosis through downregulation of Runx2. Lab Invest. 87:591–601. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Furukawa F, Matsuzaki K, Mori S, Tahashi Y, Yoshida K, Sugano Y, Yamagata H, Matsushita M, Seki T, Inagaki Y, et al: p38 MAPK mediates fibrogenic signal through Smad3 phosphorylation in rat myofibroblasts. Hepatology. 38:879–889. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Fernandez-Ramos D, Fernandez-Tussy P, Lopitz-Otsoa F, Gutiérrez-de-Juan V, Navasa N, Barbier-Torres L, Zubiete-Franco I, Simón J, Fernández AF, Arbelaiz A, et al: MiR-873-5p acts as an epigenetic regulator in early stages of liver fibrosis and cirrhosis. Cell Death Dis. 9:9582018. View Article : Google Scholar : PubMed/NCBI

39 

Zhou L, Liu S, Han M, Ma Y, Feng S, Zhao J, Lu H, Yuan X and Cheng J: miR-185 inhibits fibrogenic activation of hepatic stellate cells and prevents liver fibrosis. Mol Ther Nucleic Acids. 10:91–102. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Tao L, Xue D, Shen D, Ma W, Zhang J, Wang X, Zhang W, Wu L, Pan K, Yang Y, et al: MicroRNA-942 mediates hepatic stellate cell activation by regulating BAMBI expression in human liver fibrosis. Arch Toxicol. 92:2935–2946. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Hyun J, Park J, Wang S, Kim J, Lee HH, Seo YS and Jung Y: MicroRNA expression profiling in CCl(4)-induced liver fibrosis of Mus musculus. Int J Mol Sci. 17:E9612016. View Article : Google Scholar : PubMed/NCBI

42 

Chen W, Zhao W, Yang A, Xu A, Wang H, Cong M, Liu T, Wang P and You H: Integrated analysis of microRNA and gene expression profiles reveals a functional regulatory module associated with liver fibrosis. Gene. 636:87–95. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Murakami Y, Toyoda H, Tanaka M, Kuroda M, Harada Y, Matsuda F, Tajima A, Kosaka N, Ochiya T and Shimotohno K: The progression of liver fibrosis is related with overexpression of the miR-199 and 200 families. PloS One. 6:e160812011. View Article : Google Scholar : PubMed/NCBI

44 

Zhu J, Zhang Z, Zhang Y, Li W, Zheng W, Yu J, Wang B, Chen L, Zhuo Q, Chen L, et al: MicroRNA-212 activates hepatic stellate cells and promotes liver fibrosis via targeting SMAD7. Biochem Biophys Res Commun. 496:176–183. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 19 Issue 1

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
Zhang Y, Liu J, Ma Y, Wang J, Zhu J, Liu J and Zhang J: Integration of high‑throughput data of microRNA and mRNA expression profiles reveals novel insights into the mechanism of liver fibrosis. Mol Med Rep 19: 115-124, 2019
APA
Zhang, Y., Liu, J., Ma, Y., Wang, J., Zhu, J., Liu, J., & Zhang, J. (2019). Integration of high‑throughput data of microRNA and mRNA expression profiles reveals novel insights into the mechanism of liver fibrosis. Molecular Medicine Reports, 19, 115-124. https://doi.org/10.3892/mmr.2018.9641
MLA
Zhang, Y., Liu, J., Ma, Y., Wang, J., Zhu, J., Liu, J., Zhang, J."Integration of high‑throughput data of microRNA and mRNA expression profiles reveals novel insights into the mechanism of liver fibrosis". Molecular Medicine Reports 19.1 (2019): 115-124.
Chicago
Zhang, Y., Liu, J., Ma, Y., Wang, J., Zhu, J., Liu, J., Zhang, J."Integration of high‑throughput data of microRNA and mRNA expression profiles reveals novel insights into the mechanism of liver fibrosis". Molecular Medicine Reports 19, no. 1 (2019): 115-124. https://doi.org/10.3892/mmr.2018.9641