Hindawi BioMed Research International Volume 2017, Article ID 6484835, 17 pages https://doi.org/10.1155/2017/6484835

Research Article Expression Profiling of Cellular MicroRNA in Asymptomatic HBsAg Carriers and Chronic Hepatitis B Patients Xianliang Hou, Yan Liang, Jianing Chen, Yingfeng Wei, Ping Zeng, Lin Wang, Chong Lu, and Hongyan Diao State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China Correspondence should be addressed to Hongyan Diao; [email protected] Received 22 January 2017; Revised 31 May 2017; Accepted 22 June 2017; Published 23 August 2017 Academic Editor: Stephen H. Safe Copyright © 2017 Xianliang Hou et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Background. MicroRNAs (miRNAs) may serve as potential molecular markers to predict liver injury resulting from chronic hepatitis B (CHB). In the present study, we want to study the expression profile and clinical significance of miRNAs at different stages of CHB virus infection. Methods. Using miRNA microarray, we investigated the global expression profiles of cellular miRNA in asymptomatic hepatitis B antigen carriers (ASCs) and CHB patients, compared with healthy controls (HCs). Results. We identified 79 and 203 differentially expressed miRNAs in the peripheral blood mononuclear cells of ASCs and CHB patients compared to HCs, respectively. Some of these miRNAs were common to ASCs and CHB patients, but another set of miRNAs that showed differential expression between ASCs and CHB patients was also identified. Gene ontology and pathway enrichment analysis showed that the target genes of the identified miRNAs played a role in important biological functions, such as learning or memory, cell-cell adherens junction, ion channel inhibitor activity, TGF-beta signaling pathway, and p53 signaling pathway. Conclusion. We identified some significant differentially expressed miRNA in different phases of HBV infection, which might serve as biomarkers or therapeutic targets in the future.

1. Introduction Hepatitis B virus (HBV) is a hepatotropic noncytopathic DNA virus that is a major cause of liver diseases [1]. Eradication of HBV infection remains a global health challenge. More than 350 million people worldwide are persistent carriers of HBV, and many may progress to chronic liver disease. One to two million people die annually worldwide from HBVrelated disease [2], which results in an increase in healthcare cost and other socioeconomic burdens. In most adults, HBV infection is self-limiting and characterized by quick viral clearance; however, in some cases, the patients become carriers or develop chronic persistent infection. According to their serological profile [3], patients can be divided into two well-distinguished subsets of subjects: (1) asymptomatic HBV carriers (ASCs) and (2) chronic hepatitis B (CHB) patients. ASCs show long-lasting inhibition of viral replication with viral load levels that are usually below 2,000 IU/mL and no biochemical, ultrasonographic, or histological evidence of

liver injury. On the contrary, anti-HBe-positive CHB patients have active liver disease with a high risk of progression toward cirrhosis [4]. The difference in the responses to HBV infection is probably related to the exclusive dependence of HBV on host cellular machinery for its propagation and survival. Therefore, investigation of the interactions between HBV and host cells is crucial for understanding viral pathogenesis and the development of new antiviral therapies. MicroRNAs (miRNAs) are small noncoding RNA molecules that are about 22 nucleotides long and regulate gene expression by base pairing with the 3󸀠 -untranslated region of target mRNAs, which usually leads to mRNA degradation or translational silencing. miRNAs have been identified in most types of cells and tissues and are involved in a variety of biological processes, such as inflammation, cell proliferation, development, differentiation, apoptosis, and tumorigenesis. Further, miRNAs play vital roles in the pathogenesis of various diseases, such as cancers and viral infections, through posttranscriptional regulation of more

2 than 30% of human genes [5]. Cellular miRNAs also affect virus replication and pathogenesis, as demonstrated in the case of the liver-specific miRNA miR-122, which is essential for the replication of hepatitis C virus [6]. In addition, Zhang et al. found that the plasma miRNA profiles can indeed be used as a predictor of early virological response to interferon treatment in CHB patients [7]. In line with these findings, some reports suggest that circulating miRNAs may serve as potential molecular markers of liver injury resulting from CHB [8–10]. As the viral titer in the body, the degree of liver damage, and the immune characteristics vary between ASCs and CHBs, the expression profiles of miRNAs may also differ between these two groups of patients. However, there is not much information available about the relationships between cellular miRNAs and the different phases of chronic HBV infection. Therefore, the present study was conducted with the aim of filling in this gap in information. Using miRNA microarray and PCR analysis, we investigated the global expression profiles of cellular miRNAs in peripheral blood mononuclear cells (PBMCs) from ASCs and CHB patients and identified a few novel miRNAs that were closely involved with the pathogenesis of HBV infection. Further, network analyses were used to determine the biological roles played by the target genes of these miRNAs in both ASCs and CHB patients.

2. Material and Methods 2.1. Clinical Samples. Human blood samples were obtained from healthy donors and patients with their informed consent. The study group included sixteen ASC patients, sixteen CHB patients, and sixteen healthy controls (HCs), who were recruited from the First Affiliated Hospital, College of Medicine, Zhejiang University, between 2014 and 2015. The study protocol conforms to the ethical guidelines of the 1975 Declaration of Helsinki, and all the patients provided their written informed consent for participation. The symptoms of all the patients were diagnosed according to the previously described criteria [11]. The level of HBs antigen was measured quantitatively using the Abbott chemiluminescence immunoassay kit (Abbott Japan, Tokyo, Japan). Quantitative detection of HBV DNA was performed using an ABI7300-type quantitative PCR instrument (Applied Biosystems, USA). The results are expressed as the number of log10 international units per milliliter. 2.2. Separation of PBMCs, RNA Extraction, and miRNA Microarray. Samples containing 5 ml of blood were collected. PBMCs were isolated using standard density-gradient centrifugation on Ficoll-Paque (Amersham Biosciences, Freiburg, Germany). Total cellular RNA from the PBMCs was extracted using the TRIzol RNA reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. Low-molecular-weight RNA was isolated using the mirVana miRNA isolation kit (Ambion, Austin, TX, USA) [12]. We performed miRNA microarray analysis to identify HBVassociated differences in cellular miRNA profiles between three ASC patients and three CHB patients and three healthy controls. miRNA microarray analysis was performed using

BioMed Research International Table 1: Primer sequence of quantitative RT-PCR. Gene name U6 hsa-miR-195-3p hsa-miR-144-5p hsa-miR-451a hsa-miR-920

Primer sequence F: 5󸀠 -GCTTCGGCAGCACATATACTAAAAT-3󸀠 R: 5󸀠 -CGCTTCACGAATTTGCGTGTCAT-3󸀠 F: 5󸀠 -CCAATATTGGCTGTGCTGCTC-3󸀠 F: 5󸀠 -CGCGGATATCATCATATACTGTAAG-3󸀠 F: 5󸀠 -CGAAACCGTTACCATTACTGAGTT-3󸀠 F: 5󸀠 -GGGGAGCTGTGGAAGCAGT-3󸀠

the miRMAX microarray at KangChen Bio-Tech Corporation (Shanghai, China) [13]. The arrays were scanned using an Axon GenePix 4000B microarray scanner, and GenePix pro V6.0 was used to read the raw intensity of the image. We used the median normalization method to obtain normalized data (normalized data = [foreground value – background value]/median, where the median is the 50 percent quantile of microRNA intensity, which was greater than 30 in all the samples after background correction). An miRNA was considered to be differentially expressed if the difference in its expression was more than 2.0-fold. 2.3. Quantitative Real-Time PCR for miRNA Verification. The quantitative PCR validation group consisted of 16 CHB patients, 16 ASC patients, and 16 healthy controls. Four differentially expressed miRNA were randomly selected for verification. Expression of these mature miRNAs was assayed using the Mir-X miRNA First-Strand Synthesis Kit (number 638313, Clontech Laboratories, Inc.) and SYBR Premix Ex Taq II (Tli RNase H Plus) kit (number RR820A, Takara Bio, Inc.). Briefly, in the Mir-X cDNA synthesis reaction, RNAs are poly(A)-tailed using poly(A) polymerase and then copied using a modified oligo(dT) primer and SMART MMLV Reverse Transcriptase. The reaction mixture contained 5 𝜇l of 2× mRQ Buffer, 3.75 ul RNA sample (0.25–8 ug), and 1.25 ul mRQ Enzyme. The reaction was performed as follows: 1 hour at 37∘ C and then terminate at 85∘ C for 5 min to inactivate the enzymes. These synthetic cDNA were then specifically and quantitatively amplified using a miRNA-specific primer and SYBR advantage qPCR chemistry. All the primers for quantitative PCR (shown in Table 1) were synthesized by Sangon Biotech (Shanghai) Co., Ltd. The reaction mixture contained 5 𝜇l of 2× SYBR Premix Ex Taq II (Tli RNase H Plus), 0.2 ul miRNA-specific primer (20 uM), 0.2 ul mRQ 3󸀠 primer, 0.2 ul ROX Reference Dye (50X), and nucleasefree water, which made a total volume of 8 𝜇l. Each 8 𝜇l mixture was added to a well in a 384-well PCR plate, and this was followed by addition of 2 𝜇l cDNA in each hole. The reaction conditions were as follows: 95∘ C for 2 min followed by 40 cycles of 95∘ C for 15 s and 60∘ C for 45 s [14]. The relative amount of miRNA was normalized against an internal control, U6 snRNA, and the fold change in the amount of each miRNA was calculated using the 2−ΔΔCT method.

BioMed Research International

3 Table 2: Clinical data of patients used for cellular miRNA analysis.

Clinical characteristics

HC

ASC

CHB

𝑃 value

Number of patients

16

16

16

𝑃 = NS

6 10

5 11

6 10

𝑃 = NS

Sex number (%) Female Male Age (years) Median

45

43

42

Range

34–65

27–64

25–62

𝑃 = NS

Median

27.43

34.38

95.36

𝑃 < 0.001 (CHB versus HC)

Range

9–69

17–89

16–253

𝑃 < 0.001 (CHB versus ASC)

24

35.29

75.50

𝑃 = 0.028 (CHB versus HC )

Range HBV DNA Median Range HBsAg

14–34

21–84

9–327

𝑃 = NS (CHB versus ASC )

NA NA

1.51 × 106 2.12 × 103 –5.99 × 106

2.70 × 108 2.37 × 104 –2.51 × 109

𝑃 = NS (CHB versus ASC)

HBsAg (+) HBsAg (−)

NA NA

4 (25%) 12 (75%)

10 (62.5%) 6 (37.5%)

𝑃 = 0.033 (CHB versus ASC)

𝑃 = NS (ASC versus HC )

ALT

𝑃 = NS (ASC versus HC)

AST Median

2.4. Gene Ontology Analysis and Signaling Pathway Analysis. Target genes of the differentially expressed miRNAs were predicted based on agreement between the two databases to predict miRNAs’ target genes: targetscan7.1 and mirdbV5. Signaling pathway analysis and gene ontology (GO) analysis were performed to determine the biological functions of the target genes of differentially expressed miRNAs. The genes were mapped to each node in the GO database, and the number of genes in each node was calculated. Signaling pathway analysis is a functional analysis for mapping genes to KEGG pathways. GenMAPP v2.1 was used to map the genes to the KEGG database through the signaling pathway, and then statistical analysis was performed to determine the degree of enrichment of the genes in each pathway [15]. 2.5. Statistical Analysis. Data analysis was performed using SPSS 19. Data were expressed as the mean ± standard deviation values. The statistical significance of differences between two groups was analyzed by the 𝑡-test.

3. Results 3.1. Patient Characteristics. The clinical characteristics of the patients according to group are shown in Table 2. The mean age and male : female ratio were similar between the three groups. The distribution of ALT and AST was not found to be different between ASC group and HC group, but it is significantly increased in CHB group compared with ASC group and HC group. The average amounts of HBV DNA were higher in CHB than in ASC, but there was no significant difference. The prevalence of hepatitis B surface antigen

(HBsAg) was 62.5% in CHB and 25% in ASC. There was significant difference between them (𝑃 = 0.033). 3.2. miRNA Expression by Microarray Analysis. Among the 2077 human mature miRNAs investigated by the arrays, 79 miRNAs were found to be differentially expressed (>2-fold) between the ASC group and HC group, which accounted for 3.80% of all the miRNAs investigated. Of these, 11 were upregulated and 68 were downregulated in the ASC group. In addition, 203 miRNAs (9.77%) were differentially expressed (>2-fold) between the CHB group and HC group, including 118 upregulated miRNAs and 85 downregulated miRNAs in the CHB group. Further, 144 (6.93%) miRNAs exhibited more than a 2-fold difference in expression between the CHB group and the ASC group, of which 115 miRNAs were upregulated and 29 were downregulated. Among the differentially expressed miRNAs, 38 miRNAs were common to both the ASC group and CHB group, 5 of which were upregulated and 33 of which were downregulated in both groups (Table 3). 3.3. Validation of the Microarray Results by Quantitative Real-Time PCR. Four miRNAs with abnormal expression, namely, hsa-miR-195-3p, hsa-miR-144-5p, hsa-miR-451a, and hsa-miR-920, were selected and analyzed by real-time quantitative PCR in order to validate the microarray results. Consistent with the array data, the expression of these miRNAs was either upregulated or downregulated in the ASC group and CHB group in comparison with the HC group (Figure 1). The expression levels of hsa-miR-920 and hsamiR-195-3p still exhibited more than a 1.5-fold increase in

4

BioMed Research International Table 3: Common miRNAs differentially expressed between ASC group and CHB group. CHB group Up

Down

ASC group miR-181a-2-3p, miR-876-5p, miR-3184-3p, miR-4711-3p, miR-181b-5p

Up

miR-151a-5p, miR-330-5p, miR-130a-3p, miR-718, miRPlus-B1114, miR-326, miR-151a-3p, miR-139-5p, miR-369-3p, miR-33a-5p, miR-33b-5p, miR-3679-3p, miR-708-5p, miR-199a-3p, miR-190a-5p, miR-335-5p, miR-122-3p, miR-376a-5p, miR-495-3p, miR-498, miR-K12-8-5p, miR-22-3p, miR-126-3p, miR-1915-3p, miR-4328, miR-106b-5p, miR-331-3p, miR-151a-5p, miR-199a-5p, miR-339-5p, miR-143-3p, miR-652-3p, miR-4723-3p

Down

miR-144-5p P = 0.011

1.5

1.0

0.5

2.0 1.5 1.0 0.5 0.0

0.0 HC

ASC

CHB miR-451a P < 0.001

2.0

CHB miR-920 P = 0.011

2.5

P = 0.008 Relative expression level

NS Relative expression level

hsa-miR-195-3p P = 0.0002

2.5

Relative expression level

Relative expression level

2.0

1.5

1.0

0.5

2.0 1.5 1.0 0.5 0.0

0.0 HC

ASC

CHB

HC

CHB

Figure 1: Validation of the microarray data by qRT-PCR analysis. Changes in the expression of the four randomly selected miRNAs showed good agreement between the RT-PCR and microarray results.

the CHB group when compared to the HC or ASC group (𝑃 < 0.05). Furthermore, the expression of hsa-miR-144-5p was still significantly downregulated in the CHB group, with around 2-fold difference (𝑃 < 0.05). The expression of hsamiR-451a was similar between the HC and ASC groups, but

it was significantly higher in the CHB group (𝑃 < 0.001 and 𝑃 = 0.008, resp.). 3.4. Correlation of Four miRNAs with Clinical Indicators of ASC or CHB Patients. To investigate whether cellular

BioMed Research International

5 hsa-miR-600 hsa-miR-433-5p hsa-miR-196b-3p

2 1

hsa-miR-142-3p

0

hsa-miR-144-5p

−1

hsa-miR-130a-3p hsa-miR-199a-3p/hsa-miR-199b-3p

−2

hsa-miR-377-3p hsa-miR-654-3p hsa-miR-324-5p hsa-miR-335-3p hsa-miR-382-3p hsa-miR-559 hsa-miR-451a hsa-miR-635 hsa-miR-195-3p hsa-miR-212-3p hsa-miR-378b hsa-miR-193a-3p hsa-miR-34a-5p hsa-miR-376c-3p hsa-miR-500a-3p hsa-miR-130b-3p hsa-miR-301a-3p hsa-miR-181a-2-3p hsa-let-7b-3p hsa-miR-181b-5p hsa-miR-490-3p hsa-miR-920 CHB-3

CHB-2

CHB-1

ASC-3

ASC-2

ASC-1

HC-3

HC-2

HC-1

ebv-miR-BART7-5p

Figure 2: Hierarchical cluster analysis of the differentially expressed miRNAs. The red boxes represent upregulation of the corresponding miRNA, and the green boxes represent downregulation of the corresponding miRNA.

miR-195-3p, miR-144-5p, miR-451a, and miR-920 were related to HBV infection, we correlated these four miRNAs’ expression level with clinical indicators of HBV infection including serum ALT level, HBsAg level, and HBV DNA level. However, no significant correlation was found. Although the levels of miR-144-5p and miR-451a were slight negatively correlated with ALT (𝑟 = −0.329, −0.270; 𝑃 = 0.135, 0.135), the difference was statistically insignificant. 3.5. GO Analysis of the Target Genes of the Differentially Expressed miRNAs. The sample size was rather small, and there were individual differences within each group. Therefore, some false positive results were obtained, as some miRNAs showed abnormally high or low expression only in certain samples. Therefore, we screened more valuable miRNAs for target gene prediction, GO analysis, or signaling pathway analysis (Table 4). Hierarchical cluster analysis was performed to analyze the remaining data (Figure 2). The target sites were predicted using targetscan7.1 and mirdbV5.

The final target genes were those that were predicted by all the three miRNA prediction tools. miRNA-gene networks (Figure 3) based on the regulatory relationships between the miRNAs and their target genes were built. A total of 150 target genes were predicted for the differentially expressed miRNAs between the ASC and HC group; 119 genes, for the differentially expressed miRNAs between the CHB and HC group; and 36 genes, for the differentially expressed miRNAs between the CHB and ASC group. GO analysis consists of three components, namely, biological processes, cellular components, and molecular functions. The ten most commonly observed terms in each component were plotted to compare their differences between each group (Figure 4). The predicted target genes of the miRNAs that were differentially expressed in the ASC and CHB groups compared to the HC group were primarily involved with response to nutrient, regulation of primary metabolic process, cell part, kinase activity, and binding. Moreover, the predicted target genes of the miRNAs that were differentially expressed

6

BioMed Research International ENPP5

SLAIN1

SLC16A6

PIGA

FHDC1

FAM175B

CBX7

GJA1 ST18

DYNLL2 KCNA4

PRAMEF13

CHST1

VPS37A

PPP4R1 SPG20

NRG4

CPEB1

FBXO28

MYBL1

ACBD5

MID1IP1

TROVE2 HS3ST5 ZNF711

hsa-miR-500a-3p

hsa-miR-301a-3p

LDLR

RTCA

AP2M1

PTEN

hsa-miR-193a-3p HOXD13

CDK19 CCDC126

hsa-miR-130b-3p

CBFB

MIER1

MMGT1

SLC10A6 ZBTB5

PPARG DCAF7

GPM6A

ZMAT3

ERBB4

TGFBR2

ULK2

SYBU

CCDC6

CD69

RAP2C

FLI1 RAB9B

ZBTB18

ARHGAP12

RNF38

RP2

E2F6

TSC1

KDM2A

CAMK4

SLC39A5

HPRT1 HABP4

ACVR1 PRAMEF2

IL17RD

PAN3

DDX6

MMD

ZNF217 GNG4

SH3D19

RPS6KA5 TMEM55A NETO2

SCLY hsa-miR-181a-2-3p DYNLRB1

CUTC

MDM4 HTR2C

SLC36A1

HBEGF MZT1

DAZL

TMEM38B

PELI2

hsa-miR-376c-3p SYF2

NEUROD6 EN2

LGR4

SGPP1

CALCR

BLOC1S6 HCN3

SCN2B

ZNF268 CREBRF

KLF6

DLL1 LIMD2

NOTCH1

GMNC

NAV1

MPP2

SMIM15

RTN4RL1

HMBS

RNF34

FKBP1B VPS37B

ZNF781

hsa-miR-181b-5p MYCN

DPYSL4

KLF4

S1PR1

SATB2

E2F5

ASIC2 GABRA1

hsa-miR-34a-5p

CCNE2 NRIP3

hsa-miR-195-3p

FAM76A

AMER1

GABRA3

ERGIC1

GSKIP ARSJ

SYT1

PPP2R3A

NAV3

FAM167A

FLOT2

CADM2

OSBPL3 SRSF7

PVRL1

VAMP2

ZNF594 MET

TGIF2

PDGFRA

PNOC

ELMOD1

RRAS

CA7

VAT1

HNF4A

CUEDC1

JAKMIP1

BRINP1

FGD6

LEF1

(a) FOXN2

TMEM128 TAOK1

BLOC1S6

TRIM63

ZNF268 CREBRF

KLF6 IRX3

POLR2K

HMBS

RNF34

hsa-let-7b-3p

ZNF781

hsa-miR-181b-5p S1PR1

DCUN1D4

GABRA1 ARID4B

XIAP

ARMC8

GSKIP ARSJ

EIF4ENIF1

CARTPT

CCDC126 ENPP5

PITX2 RAP2C

ULK2 MMGT1

DDX6

LDLR

ZNF217

CUL1 hsa-miR-377-3p

ZMAT3 FBXO28

MDM4

OCLN

HS3ST5

ACVR1

SH3D19

OSBPL3

ZNF594

FAM175B

DCP1A

ACBD5

HABP4 TGFBR2

TSC1

C5orf24

MIER1

CD69

MID1IP1

TMEM55A MYBL1 RPS6KA5 SYBU CPEB1

CBFB

SLC6A19

KCNA4

PIGA

PAN3

FOXJ3

ZBTB18

hsa-miR-130a-3p

SLAIN1 TROVE2 ARHGAP12

CHST1

RNF38

KDM2A RTCA

DYNLL2

VPS37A

ST18

CDK19 SPG20 GJA1

PTEN

PPARG

SLC16A1

OSR1

hsa-miR-144-5p

hsa-miR-451a

RAB33A

HPRT1 hsa-miR-654-3p

GMFB

hsa-miR-600

SLC2A10 CCDC53

GNG4

HTR2C

RGR

UBE2A

VDAC1

GNAI2 CUTC

DYNLRB1

SRP9

DUSP23

MZT1 DAZL hsa-miR-181a-2-3p

MGAT3

LAMTOR3

SMCR8 SCLY

RALA

hsa-miR-559

CAMKV

hsa-miR-324-5p

hsa-miR-920

MMD

ATPAF1 RAB9B

AK3 RBPMS2

MGARP

SLC36A1 FLI1

PELI2

LIMD2

OR7D2

NETO2

(b)

Figure 3: Continued.

TMCC1 PCYT1B

SRSF7

BioMed Research International

7 FOXJ3

EIF4ENIF1

RASA1

GJD4 RGS7BP

PITX2

XIAP

LEMD3 PTPRM PAIP2

CARTPT CUL1

hsa-miR-635 C8orf44-SGK3

hsa-miR-377-3p

hsa-miR-212-3p

HS3ST5 CD69

ARMC8 SAP30L OCLN

CALU

NAA15

RNF38 C5orf24

DCP1A

KCNK2

TIMM9

SLC6A19

SLC16A1

VDAC1

hsa-miR-654-3p

hsa-miR-490-3p

GMFB

TMOD3

OSR1

SIRT7

SDR9C7

hsa-miR-451a

hsa-miR-433-5p

hsa-miR-196b-3p

FUT9

hsa-miR-335-3p

hsa-miR-195-3p

CADM2

GOLT1A

hsa-miR-378b

(c)

Figure 3: miRNA-gene networks built using the target genes of the differentially expressed miRNAs between the ASC group and HC group (a), CHB group and HC group (b), and CHB group and ASC group (c). The red squares represent miRNAs, and the blue round spots represent genes. The lines represent the interactions between miRNAs and genes.

Table 4: Significant differentially expressed miRNA in each group.

Upregulation

Downregulation

ASC versus HC

CHB versus HC

hsa-miR-181a-2-3p, hsa-miR-142-3p hsa-miR-181b-5p

hsa-miR-181a-2-3p hsa-miR-920 hsa-let-7b-3p hsa-miR-181b-5p

hsa-miR-376c-3p hsa-miR-199a-3p hsa-miR-130b-3p hsa-miR-193a-3p hsa-miR-195-3p hsa-miR-500a-3p hsa-miR-34a-5p hsa-miR-301a-3p

hsa-miR-130a-3p hsa-miR-382-3p hsa-miR-559 hsa-miR-377-3p hsa-miR-654-3p hsa-miR-451a hsa-miR-600 hsa-miR-144-5p hsa-miR-324-5p

between the CHB group and ASC group were involved with learning or memory, cell-cell adherens junction, and ion channel inhibitor activity. Pathway enrichment analysis was performed to further understand the functions and signal pathways of these predicted gene targets (Figure 5). The results indicated that the target genes of differentially expressed miRNAs between the ASC group and HC group were related to 26 signaling pathways, particularly the pathways associated with endocytosis, thyroid cancer, and p53 signaling pathway. Further, the target genes of the differentially expressed miRNAs between the CHB group and HC group played a role in 12 signaling pathways, primarily the TGF-beta signaling pathway, p53 signaling pathway, and RNA degradation. Finally, the target genes of the differentially expressed miRNAs between the ASC group and CHB group were related to 4 signaling pathways that were primarily involved with TGF-beta, ubiquitin mediated proteolysis, and cell adhesion molecules (CAMs).

CHB versus ASC hsa-miR-490-3p hsa-miR-212-3p hsa-miR-635 ebv-miR-BART7-5p hsa-miR-195-3p hsa-miR-378b hsa-miR-335-3p hsa-miR-433-5p hsa-miR-377-3p hsa-miR-654-3p hsa-miR-451a hsa-miR-196b-3p

3.6. miRNA-GO/Pathway Network Analysis. To better understand the associations between the differentially expressed miRNAs and the results of GO and signaling pathway analyses, miRNA-GO networks (Figure 6) and miRNA-pathway networks (Figure 7) were built. Differentially expressed miRNAs, the GO terms that their target genes were linked to, and the pathways they were involved in according to the pathway enrichment analysis are represented as nodes in the graph, and the biological relationship between two nodes is represented as a line. All lines were supported by at least one study from the published literature, a textbook, or functional information in the GO or KEGG database. Based on the lines, we could then infer the key roles of the relationships depicted in the networks. The degree of complexity of each node was of primary concern, because the weight (importance) of the regulator (in this case, the miRNA) increases as the degree of complexity increases, according to the theory of network biology [24]. The visualized networks indicated that

Behavior Learning or memory Single-organism behavior Cognition Embryonic hindlimb morphogenesis Response to food Hindlimb morphogenesis Regulation of actin filament polymerization Negative regulation of biological process Actin filament polymerization Cell-cell junction Cytoplasmic mRNA processing body N-terminal protein acetyltransferase complex Cell-cell adherens junction Cytoplasm Cell part Integral component of nuclear inner membrane Intrinsic component of nuclear inner membrane Cell Intrinsic component of the cytoplasmic side of the plasma membrane Potassium channel regulator activity Channel regulator activity Ion channel inhibitor activity Channel inhibitor activity Histone deacetylase activity Protein deacetylase activity mRNA binding Deacetylase activity Ubiquitin protein ligase activity Ubiquitin-like protein ligase activity

Enrichment score (− FIA 10(P value))

5.00

4.00

3.00

2.00

1.00 Enrichment score (− FIA 10(P value))

6.00

0.00

Biological process Cellular component Molecular function

Biological process Cellular component

Negative regulation of cardiac muscle cell proliferation Response to nutrient levels Regulation of cellular metabolic process Regulation of metabolic process Muscle tissue development Negative regulation of cardiac muscle tissue growth Negative regulation of heart growth Response to extracellular stimulus Response to nutrient Regulation of primary metabolic process Cytoplasmic mRNA processing body Intracellular Intracellular part Cell part Cell Intracellular membrane-bounded organelle Intracellular organelle Whole membrane Cytoplasmic ribonucleoprotein granule Late endosome Ubiquitin protein ligase activity Ubiquitin-like protein ligase activity Transferase activity Acetylglucosaminyltransferase activity Transmembrane receptor protein serine/threonine kinase activity Transforming growth factor beta binding GDP binding mRNA binding Translation regulator activity, nucleic acid binding Ubiquitin-protein transferase activity

Positive regulation of cellular process Generation of neurons Neurogenesis Neuron differentiation Cell development Muscle cell proliferation Regulation of cardiac muscle cell proliferation Regeneration Cardiac muscle tissue growth Positive regulation of cellular biosynthetic process Cell part Cell Intrinsic component of plasma membrane Whole membrane Plasma membrane part Integral component of plasma membrane Bounding membrane of organelle Clathrin-coated vesicle membrane Synapse Escrt I complex Syntaxin-1 binding Transmembrane receptor protein kinase activity Phosphatidylinositol-4,5-bisphosphate 3-kinase activity Phosphatidylinositol bisphosphate kinase activity Protein kinase activity Transcription factor activity, sequence-specific DNA binding Phosphatidylinositol 3-kinase activity Nucleic acid binding transcription factor activity Metal ion transmembrane transporter activity Transcription factor activity, transcription factor recruiting

Enrichment score (− FIA 10(P value))

8 BioMed Research International

5.00

4.50

4.00

3.50

3.00

2.50

2.00

1.50

1.00

0.50

0.00

Biological process Cellular component

(a)

Molecular function

4.50

(b)

4.00

3.50

3.00

2.50

2.00

1.50

1.00

0.50

0.00

Molecular function

(c)

Figure 4: Functional classification of the target genes of differentially expressed miRNAs between the ASC group and HC group (a), CHB group and HC group (b), and CHB group and ASC group (c).

10 9 8 7 6 5 4 3 2 1 0

Enrichment score Gene counts

3.00 2.50 2.00 1.50 1.00 0.50 0.00

TGF-beta signaling pathway: Homo sapiens (human) p53 signaling pathway: Homo sapiens (human) RNA degradation: Homo sapiens (human) Pathways in cancer: Homo sapiens (human) mTOR signaling pathway: Homo sapiens (human) Gap junction: Homo sapiens (human) GABAergic synapse: Homo sapiens (human) Morphine addiction: Homo sapiens (human) Longevity regulating pathway: Homo sapiens (human) Circadian entrainment: Homo sapiens (human)

4.00 3.50 3.00 2.50 2.00 1.50 1.00 0.50 0.00

9

Endocytosis: Homo sapiens (human) Thyroid cancer: Homo sapiens (human) p53 signaling pathway: Homo sapiens (human) TGF-beta signaling pathway: Homo sapiens (human) Pathways in cancer: Homo sapiens (human) Prostate cancer: Homo sapiens (human) Longevity regulating pathway: Homo sapiens (human) MicroRNAs in cancer: Homo sapiens (human) Synaptic vesicle cycle: Homo sapiens (human) Transcriptional misregulation in cancer: Homo sapiens (human)

BioMed Research International 8 7 6 5 4 3 2 1 0

Enrichment score Gene counts (a)

(b)

2.0

1.50

1.5

1.00

1.0

0.50

0.5

0.00

0.0 Cell adhesion molecules (CAMs): Homo sapiens (human)

NOD-like receptor signaling pathway: Homo sapiens (human)

2.00

Ubiquitin mediated proteolysis: Homo sapiens (human)

2.5

TGF-beta signaling pathway: Homo sapiens (human)

2.50

Enrichment score Gene counts (c)

Figure 5: Pathway enrichment analysis of the target genes of differentially expressed miRNAs between the ASC group and HC group (a), CHB group and HC group (b), and CHB group and ASC group (c).

miR-142-3p, miR-181b-5p, miR-199a-3p, miR-130b-3p, miR34a-5p, miR-301a-3p, and miR-376c-3p played regulatory roles in modulating the molecular networks in ASCs. Further, the networks demonstrated that miR-181b-5p, miR-130a3p, miR-559, miR-920, miR-377-3p, miR-654-3p, miR-451a, miR-600, and miR-324-5p might be crucial regulators of pathogenesis in CHB patients. Finally, it was shown that miR-212-3p, miR-490-3p, miR-635, miR-377-3p, miR-6543p, and miR-451a played a prominent role in the global signaling networks and pathways involved in the progression of chronic liver disease, as these miRNAs carried considerable weight in both the ASC and CHB groups.

4. Discussion In this report, utilizing miRNA array, we analyzed the global miRNA expression profiles in the PBMCs of healthy control individuals and HBV-infected patients who were asymptomatic carriers or had chronic hepatitis B infection. Bioinformatics analysis of the microarray results revealed a set of miRNAs that were differentially expressed in the PBMCs of ASCs and CHB patients compared to the HCs. In addition, a significant number of the differentially expressed miRNAs were found in both ASCs and CHB patients. Moreover, we found that 144 mRNAs exhibited more than a

10

BioMed Research International Organic cyclic compound metabolic process

Nucleobase-containing compound metabolic process

Cellular nitrogen compound metabolic process

Regulation of biosynthetic process

Cellular aromatic compound metabolic process

Cell communication Regulation of nucleobase-containing compound metabolic process

Biosynthetic process

Organic substance biosynthetic process

Regulation of nitrogen compound metabolic process

Heterocycle metabolic process Nitrogen compound metabolic process

Single organism signaling

Regulation of cellular metabolic process

Single-organism process

Primary metabolic process

Macromolecule biosynthetic process

Macromolecule metabolic process

Cellular metabolic process

Regulation of primary metabolic process

Response to stimulus

hsa-miR-376c-3p

hsa-miR-142-3p Regulation of metabolic process

Organic substance metabolic process

Cellular biosynthetic process

Metabolic process Biological regulation Cellular macromolecule metabolic process

Single-organism Cellular process cellular process hsa-miR-301a-3p hsa-miR-199a-3p

Signaling

Regulation of cellular process

hsa-miR-130b-3p

Central nervous system neuron development

Regulation of biological process

hsa-miR-181b-5p

hsa-miR-34a-5p hsa-miR-193a-3p

Central nervous system neuron differentiation

Regulation of peptidyl-serine phosphorylation

Response to hydrogen peroxide

Cell fate commitment

(a) Neural crest cell differentiation

Regulation of fat cell differentiation Cytokine metabolic process

Cellular process involved in reproduction in multicellular organism

Cytokine biosynthetic process

Fat cell differentiation

Sexual reproduction

Mesenchyme development

Negative regulation of biological process

Receptor metabolic process Regulation of nitrogen compound metabolic process

Regulation of transcription, DNA-templated

Response to chemical

hsa-miR-920

Cellular protein modification process

Nucleic acid metabolic process

Regulation of cellular biosynthetic process

Biosynthetic process

Organic cyclic compound biosynthetic process

Nucleobase-containing compound biosynthetic process Macromolecule biosynthetic process

Cellular protein metabolic process

RNA metabolic process

hsa-miR-654-3p

Cellular biosynthetic process

Nitrogen compound metabolic process

Positive regulation of protein binding

B cell proliferation

hsa-miR-324-5p

Heterocycle biosynthetic process Regulation of macromolecule biosynthetic process

hsa-miR-130a-3p

Organic substance metabolic process

Gene expression Regulation of cellular metabolic process

Regulation of protein binding

Macromolecule metabolic Single-organism process cellular process Cellular process

Cellular metabolic process

Primary metabolic process

Cellular macromolecule metabolic process

Regulation of rna metabolic process

Single-organism process

Embryonic epithelial tube formation

hsa-miR-181b-5p

Neuron-neuron synaptic transmission

Positive regulation of binding

Regulation of Biological cellular process regulation

Regulation of biological process

hsa-miR-451a hsa-miR-559 Regulation of gene expression

Protein metabolic process

Negative regulation of cellular process

Protein modification process

Organic substance biosynthetic process

Regulation of macromolecule metabolic process

Transcription, DNA-templated

Cellular nitrogen compound biosynthetic process

Metabolic process

B cell activation Tube formation

Regulation of biosynthetic process

Regulation of cellular macromolecule biosynthetic process

Cellular nitrogen compound metabolic process

Nucleobase-containing compound metabolic process

Regulation of nucleobase-containing compound metabolic process Macromolecule modification

RNA biosynthetic process Response to organic substance

Cellular macromolecule biosynthetic process

Response to oxygen levels

hsa-miR-600

Epithelial tube formation

Regulation of metabolic process

Heterocycle metabolic process

Aromatic compound biosynthetic process

hsa-miR-377-3p

Organic cyclic compound metabolic process

Cellular aromatic compound metabolic process

Regulation of rna biosynthetic process

Response to stimulus Multicellular organismal process

Single organism signaling

Regulation of primary metabolic process Cellular response to oxidative stress

Cell communication Signaling

(b)

Figure 6: Continued.

BioMed Research International

11 Neuron death Neuron projection morphogenesis

Cellular response to fibroblast growth factor stimulus

Regulation of cellular component organization

Neuron apoptotic process

Pallium development

Cell morphogenesis involved in differentiation

Axonogenesis RNA processing RNA 3 -end processing

Regulation of cellular metabolic process

Telencephalon development

Forebrain development

Response to fibroblast growth factor

Protein metabolic process

Regulation of gene expression Biosynthetic process

ncRNA processing

Regulation of cellular biosynthetic process

hsa-miR-212-3p Transmembrane receptor protein serine/threonine kinase signaling pathway

Response to stimulus Gene expression Axon development

hsa-miR-654-3p hsa-miR-635

Positive regulation of MAP kinase activity

Regulation of cellular response to stress

Negative regulation of cellular component organization

Positive regulation of phosphorus metabolic process

Regulation of kinase activity

Learning or memory

Regulation of biosynthetic process Regulation of RNA biosynthetic process

Organic cyclic compound biosynthetic process

Cellular protein modification process

Organic substance biosynthetic process

Nucleobase-containing Aromatic compound compound biosynthetic biosynthetic process process Cellular macromolecule biosynthetic process

RNA biosynthetic process

Cellular aromatic compound metabolic process

Positive regulation of protein phosphorylation Chromatin organization

Cellular response to oxidative stress

hsa-miR-490-3p

Positive regulation of phosphate metabolic process

Regulation of Negative protein regulation of modification kinase activity process Positive regulation of catalytic activity Positive Chromosome regulation of organization cellular protein Protein import metabolic process Chromatin modification Positive Positive regulation of regulation of protein metabolic cellular catabolic process process

Embryonic organ morphogenesis

Embryonic organ development

Transcription, DNA-templated Regulation of nucleobase-containing compound metabolic Response to process chemical

Nitrogen compound metabolic process

Nucleobase-containing compound metabolic process

Macromolecule biosynthetic process

Organic cyclic compound metabolic process

Regulation of RNA metabolic process

Cellular biosynthetic process

Cellular protein metabolic process

Macromolecule modification Single-organism behavior

Protein dephosphorylation

Cellular nitrogen compound biosynthetic process

Nucleic acid metabolic process

Response to organic substance

hsa-miR-377-3p Behavior

Negative regulation of biological process

Heterocycle metabolic process

Cellular nitrogen compound metabolic process

RNA metabolic process

Regulation of primary metabolic process

Regulation of MAP kinase activity

Regulation of cellular macromolecule biosynthetic process

hsa-miR-451a

Metabolic process

Regulation of protein kinase activity

Regulation of macromolecule biosynthetic process

Regulation of transcription, DNA-templated

Regulation of macromolecule metabolic process

Regulation of biological process Macromolecule metabolic Regulation of process cellular process Regulation of Cellular metabolic Biological macromoleculeOrganic process regulation metabolic substance process metabolic process Cellular metabolic Primary metabolic process process

Negative regulation of protein kinase activity

Heterocycle biosynthetic process

Negative regulation of cellular process

Regulation of nitrogen compound metabolic process

Protein modification process

Negative regulation of protein modification process Positive regulation of protein modification process

Regulation of MAPK cascade

Response to oxidative stress

(c)

Figure 6: miRNA-GO networks based on the identified GO terms of the target genes of the differentially expressed miRNAs between the ASC group and HC group (a), CHB group and HC group (b), and CHB group and ASC group (c). The red boxes represent differentially expressed miRNAs; the blue circles represent the significant GO terms; and the straight lines represent interactions between the miRNA and the GO term. GO, gene ontology; miRNA, microRNA.

2-fold difference in their expression between the CHB group and ASC group. These observations indicated that common and phase-specific mechanisms may exist at the miRNA level in ASCs and CHB patients. Thus, investigating these miRNAs and their mechanisms might be of great importance for further research into the pathogenesis of hepatitis B infection. miRNAs are likely to play a prominent role in altering the global signaling networks and pathways involved in the progression of liver disease. Therefore, in this study, we identified the target genes of the differentially expressed miRNAs and built miRNA-gene networks to study their interactions; further, we used GO and signaling pathway analysis to determine the functions and biological pathways of the target genes. GO analysis demonstrated that the target genes were primarily involved with response to nutrient, regulation of primary metabolic process, cell part, kinase activity, and binding, and pathway enrichment showed that the target genes were involved in several important pathways, such as those related to endocytosis, ubiquitin mediated proteolysis, TGF-beta signaling pathway, and p53 signaling pathway. Finally, the significance of the identified miRNAs was determined by building miRNA-GO and miRNA-pathway networks and calculating the degree of complexity of the nodes [57]. Using this analytical approach, we identified the

key miRNAs at the centre of the signal transduction network in patients with HBV infection (both carriers and those with chronic infection). Since the remaining miRNAs also had a considerable degree of complexity, they will be the subject of our future studies. Meanwhile, our current results indicate that miR-142-3p, miR-181b-5p, miR-199a-3p, miR-130b-3p, miR-34a-5p, miR-301a-3p, and miR-376c-3p may be closely related to disease pathogenesis in ASCs and that miR-181b-5p, miR-130a-3p, miR-559, miR-920, miR-377-3p, miR-654-3p, miR-451a, miR-600, and miR-324-5p may play an important role in the pathogenesis of chronic hepatitis B. Further, miR212-3p, miR-490-3p, and miR-635, and especially miR-3773p, miR-654-3p, and miR-451a, may be associated with the progression of hepatitis B-related diseases, as they were found to be significant in both ASCs and CHB patients. Several of these miRNAs have already been reported in previous studies, namely, miR-181b-5p, miR-199a-3p, miR-130b-3p, miR-34a5p, miR-301a-3p, miR-130a-3p, miR-654-3p, miR-451a, miR377-3p, and miR-600. However, miR-142-3p, miR-376c-3p, miR-559, miR-920, miR-324-5p, miR-212-3p, miR-490-3p, and miR-635 have not been previously reported in studies on HBV-related diseases, which makes this finding novel. No significant correlation was found between the levels of miR-195-3p, miR-144-5p, miR-451a, and miR-920 and clinical

12

BioMed Research International Choline metabolism in cancer: Homo sapiens (human)

Bacterial invasion of epithelial cells: Homo sapiens (human)

Regulation of actin cytoskeleton: Homo sapiens (human)

Adherens junction: Homo sapiens (human)

VEGF signaling pathway: Homo sapiens (human)

Non-alcoholic Hippo signaling fatty liver disease pathway: Homo (NAFLD): Homo sapiens (human) sapiens (human) Signaling pathways regulating pluripotency of Renal cell stem cells: Homo carcinoma: sapiens (human) Homo sapiens (human)

hsa-miR-199a-3p hsa-miR-142-3p

Fc gamma R-mediated phagocytosis: Homo sapiens (human) AGE-RAGE signaling pathway in diabetic complications: Homo sapiens (human)

MAPK signaling pathway: Homo sapiens (human)

Proteoglycans in cancer: Homo sapiens (human)

Fc epsilon RI signaling pathway: Homo sapiens (human)

hsa-miR-301a-3p

Neurotrophin signaling pathway: Homo sapiens (human)

Sphingolipid signaling pathway: Homo sapiens (human)

Endocytosis: Homo sapiens (human)

hsa-miR-130b-3p

Ras signaling pathway: Homo sapiens (human)

Estrogen signaling pathway: Homo sapiens (human)

hsa-miR-34a-5p Axon guidance: Homo sapiens (human) hsa-miR-181b-5p Transcriptional misregulation in cancer: Homo sapiens (human)

Glycosaminoglycan biosynthesis, keratan sulfate: Homo sapiens (human)

TGF-beta signaling pathway: Homo sapiens (human)

Amino sugar and nucleotide sugar metabolism: Homo sapiens (human)

T cell receptor signaling pathway: Homo sapiens (human) Glycosaminoglycan biosynthesis, heparan sulfate/ heparin: Homo sapiens (human)

Protein processing in endoplasmic reticulum: Homo sapiens (human)

(a) Glycosaminoglycan biosynthesis, heparan sulfate/ heparin: Homo sapiens (human)

Estrogen signaling pathway: Homo sapiens (human) Axon guidance: Homo sapiens (human) FoxO signaling pathway: Homo sapiens (human)

hsa-miR-181b-5p

TGF-beta signaling pathway: hsa-miR-130a-3p

Transcriptional

Homo sapiens (human)

misregulation in cancer: Homo sapiens (human)

Neurotrophin signaling pathway: Homo sapiens (human)

MAPK signaling pathway: Homo sapiens (human)

hsa-miR-559

(b)

Figure 7: Continued.

BioMed Research International

13 Hippo signaling Adherens junction:

pathway: Homo

Homo sapiens

sapiens (human)

(human)

Colorectal cancer: TGF-beta

Homo sapiens

signaling pathway:

(human)

Homo sapiens (human)

Endometrial cancer: Homo Axon guidance:

sapiens (human)

Homo sapiens (human)

hsa-miR-212-3p

Thyroid cancer: Homo sapiens

Oocyte meiosis:

(human)

Homo sapiens (human)

Basal cell carcinoma:

Cell cycle: Homo

Homo sapiens

sapiens (human) Glutamatergic

(human)

synapse: Homo sapiens (human)

(c)

Figure 7: miRNA-pathway networks based on the identified pathways of the target genes of the differentially expressed miRNAs between the ASC and HC group (a), between the CHB and HC group (b), and between the CHB and ASC group (c). The red squares represent miRNAs; the blue round spots represent the pathways; and the lines represent interactions between the miRNAs and their target gene pathways.

Table 5: Correlation of miRNAs and clinical indicators of HBV infection. MicroRNA

Relevance

Clinical indicators

Citation

Positive correlation

HBV DNA

[16]

Positive correlation Positive correlation Positive correlation

ALT levels HBsAg levels, ALT levels, and HBV DNA titers HBsAg levels, ALT levels

[17] [18] [18]

miR-29

Positive correlation Negatively correlated

HBV DNA Liver fibrotic stages and necroinflammation grades

[16] [19]

miR-210 miR-33a miR-125b

Positive correlation Positive correlation Negatively correlated

HBV DNA, HBs antigen, alanine aminotransferase (ALT), and HAI score Hepatic fibrosis HBV DNA intermediates and secretion of HBsAg and HBeAg

[20] [21] [22]

miR-146a

Positive correlation Positive correlation

HBsAg levels ALT

[23] [14]

miR-548ah-5p

Negative correlation

HBV DNA

[14]

miR-122 miR-22

indicators of HBV infection. It is likely due to the small sample size of this study and clinical information missed in some patients. Taking the correlated between miR-144-5p, miR-451a, and ALT as an example, it was possible that the association was real; with a larger sample, we might find that the association did become statistically significant. In

attempts to answer these questions, we summarized miRNAs that was reported as a strong correlation with clinical indicators of HBV infection (Table 5). The functions of the previously reported miRNAs are described here. Previous studies have demonstrated that miRNA-181a is a critical player in the modulation of both

14

BioMed Research International Table 6: Deregulated miRNA in HBV infection or HBV-related disease.

MicroRNA miR-1 miR-15a miR-15b miR-15a/16 miR-17 miR-17-92 cluster miR-18a miR-19b miR-20a miR-21 miR-22 miR-26b miR-27a miR-29a miR-29c miR-33a miR-34c miR-93 miR-99a miR-101 miR-103 miR-106a miR-107 miR-122 miR-125a-5p miR-125b miR-125b-5p miR-126 miR-130a miR-141 miR-143 miR-145 miR-146a miR-148a miR-152 miR-155 miR-181a miR-199a miR-205 miR-210 miR-221 miR-224 miR-331-3p miR-370 miR-372/373 miR-429 miR-449a

Alteration

Target

Citation

Up Down Down (early stage) Up (late stage) Down Up Up Up Up Up Up Down Down Up Up Down Up Down Down Down Down Up Up Up Down Up Down Up Down Down — Up Down Up Up Down Up Up Up Down Up Up Up Up Down Up Down Up

HDAC4, FXRA HBp and HBx

[25] [26]

HNF1𝛼

[27]

Bcl-2 ULK1, ATG7, p62 HBV transcripts, E2F1 ESR1, ERa P53 Egln3/PHD3 PDCD4, PTEN CDKN1A, AKT3, p21 CHORDC1 MAP2K4, TR𝛽1 PTEN TNFAIP3 Smad7 TGIF2 MICA AGO2 FOXO1, EZH2, DNMT3A PER3, CDK5R1 P130, FAS CDK8, let-7 HBV DNA polymerase, Cyclin G1, HO-1, NDRG3, PPAR𝛾, PBF HBsAg SCNN1A HBsAg, LIN28B/let-7 axis PI3KR2, Crk, PLK2 PPARG, Era, PGC1𝛼, PPAR𝛾 PPAR𝛼 FNDC3B CUL5 STAT1, CFH, RIG-I, RIG-G c-Met, Wnt DNMT1 C/EBP, SOCS1 HLA-A, E2F5 HBsAg HBx HBV pre-S1 PI3-K/Akt HOXD10, CDC42, Smad4 ING5 NFIA NFIB NOTCH1 CREBFXRA

[28] [29] [30, 31] [29, 32] [30] [30] [33] [34, 35] [36] [30] [37] [38] [21] [39] [40] [41] [42–44] [29] [29] [29] [45–47] [46, 48] [22] [49, 50] [30] [30, 45, 51] [46] [47] [52] [23, 46, 53] [30] [46] [46] [46, 47] [46] [46] [46] [29] [29, 47] [54] [45] [46] [55] [45]

BioMed Research International

15 Table 6: Continued.

MicroRNA miR-501 miR-545/374a miR-548ah miR-581 miR-602 miR-939 miR-1231 miR-4717 Let-7a

Alteration

Target

Citation

Up Up Up Down Down Down Up Down Down

HBXIP ESRRG INF𝛾R1 Dicer, EDEM1 RASSF1A CEBPA HBcAg PD-1 CCR7

[46] [29] [46] [46] [29] [45] [46] [56] [29]

innate [58] and adaptive [59] immunity and is involved in distinct pathological processes. miR-199a-3p has been reported to bind to and directly suppress HBV RNA [60]. miR-130a and miR-130b share the same seed sequences, and miRNA-130a can inhibit hepatitis B virus replication by targeting PGC1𝛼 and PPAR𝛾 [51]. Moreover, downregulation of miR-377 contributes to IRX3 deregulation in hepatocellular carcinoma [61]. miRNA-34is associated with hepatic fat metabolism [62]. A recent study has shown that the expression of miR-301 in the serum was significantly higher in HCC patients than in healthy subjects [63]. Finally, hasmiR-600 was found to be associated with inflammatory and cell cycle pathways [12]. In general, miRNAs could act as a cellular anti-viral defense, as siRNAs do in plants and lower eukaryotes, or cellular miRNAs could be exploited by the virus to help establish a favorable environment for its replication and survival. As shown in Table 6, we summarized miRNAs that was reported as a dysregulated miRNA by HBV infection. In summary, we have comprehensively analyzed the cellular miRNA profiles of HBV-infected carriers and chronic disease patients. We have identified certain important miRNAs that were differentially expressed in the patients and were also differentially expressed between the carriers and chronic disease patients. Further, network biology analysis of the target genes of these miRNAs demonstrated the global regulation of signaling network pathways by these miRNAs. Further studies on the molecular regulating mechanisms of miRNAs in HBV-related diseases need to be conducted to further elucidate the pathogenesis of chronic hepatitis B and identify therapeutic targets for treatment.

Conflicts of Interest The authors have stated explicitly that there are no conflicts of interest in connection with this article.

Authors’ Contributions Hongyan Diao and Xianliang Hou conceived, designed, and performed most of the experiments with significant contributions from Yan Liang, Jianing Chen, Yingfeng Wei, Ping Zeng, Lin Wang, and Chong Lu; Yan Liang, Yingfeng Wei,

and Chong Lu contributed to sample collection; Xianliang Hou, Jianing Chen, Lin Wang, and Ping Zeng analyzed the data; Hongyan Diao and Xianliang Hou wrote the paper. All authors provided final approval and agree to be accountable for all aspects of the work.

Acknowledgments This work was supported by the National Natural Science Foundation of China (nos. 81271810, 81571953), the National Basic Research Program (2013CB531405), Zhejiang Provincial Natural Science Foundation of China (LY16H190002), and Zhejiang Medical Science and Technology Project (2015118507).

References [1] C. Seeger and W. S. Mason, “Hepatitis B virus biology,” Microbiology and Molecular Biology Reviews, vol. 64, no. 1, pp. 51–68, 2000. [2] Y. W. Huang, K. Chayama, J. H. Kao, and S. S. Yang, “Detectability and clinical significance of serum hepatitis B virus ribonucleic acid,” Hepatobiliary Surgery and Nutrition, vol. 4, no. 3, pp. 197–202, 2015. [3] S. J. Hadziyannis and G. V. Papatheodoridis, “Hepatitis B e antigen-negative chronic hepatitis B: Natural history and treatment,” Seminars in Liver Disease, vol. 26, no. 2, pp. 130–141, 2006. [4] S. Maimone, G. Caccamo, G. Squadrito et al., “A combination of different diagnostic tools allows identification of inactive hepatitis B virus carriers at a single time point evaluation,” Liver International, vol. 37, no. 3, pp. 362–368, 2017. [5] B. P. Lewis, C. B. Burge, and D. P. Bartel, “Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets,” Cell, vol. 120, no. 1, pp. 15–20, 2005. [6] C. L. Jopling, M. Yi, A. M. Lancaster, S. M. Lemon, and P. Sarnow, “Molecular biology: modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA,” Science, vol. 309, no. 5740, pp. 1577–1581, 2005. [7] X. Zhang, C. Chen, M. Wu et al., “Plasma microRNA profile as a predictor of early virological response to interferon treatment in chronic hepatitis B patients,” Antiviral Therapy, vol. 17, no. 7, pp. 1243–1253, 2012.

16 [8] Y. Murakami, H. Toyoda, T. Tanahashi et al., “Comprehensive miRNA expression analysis in peripheral blood can diagnose liver disease,” PLoS ONE, vol. 7, no. 10, Article ID e48366, 2012. [9] Y. Chen, L. Li, Z. Zhou, N. Wang, C.-Y. Zhang, and K. Zen, “A pilot study of serum microRNA signatures as a novel biomarker for occult hepatitis B virus infection,” Medical Microbiology and Immunology, vol. 201, no. 3, pp. 389–395, 2012. [10] H. Zhang, Q. Y. Li, Z. Z. Guo et al., “Serum levels of mi-croRNAs can specifically predict liver injury of chronic hepatitis B,” World Journal of Gastroenterology, vol. 18, no. 37, pp. 5188–5196, 2012. [11] A. S. F. Lok and B. J. McMahon, “Chronic hepatitis B,” Hepatology, vol. 45, no. 2, pp. 507–539, 2007. [12] X.-B. Pan, H. Ma, Q. Jin, and L. Wei, “Characterization of microRNA expression profiles associated with hepatitis B virus replication and clearance in vivo and in vitro,” Journal of Gastroenterology and Hepatology, vol. 27, no. 4, pp. 805–812, 2012. [13] C.-J. Guo, Q. Pan, D.-G. Li, H. Sun, and B.-W. Liu, “miR-15b and miR-16 are implicated in activation of the rat hepatic stellate cell: an essential role for apoptosis,” Journal of Hepatology, vol. 50, no. 4, pp. 766–778, 2009. [14] T. Xing, H. Xu, W. Yu, B. Wang, and J. Zhang, “Expression profile and clinical significance of miRNAs at different stages of chronic hepatitis B virus infection,” International Journal of Clinical and Experimental Medicine, vol. 8, no. 4, pp. 5611–5620, 2015. [15] Y.-F. Pan, T. Qin, L. Feng, and Z.-J. Yu, “Expression profile of altered long non-coding RNAs in patients with HBV-associated hepatocellular carcinoma,” Journal of Huazhong University of Science and Technology—Medical Science, vol. 33, no. 1, pp. 96– 101, 2013. [16] T. J. Xing, D. F. Jiang, J. X. Huang, and Z. L. Xu, “Expression and clinical significance of miR-122 and miR-29 in hepatitis B virusrelated liver disease,” Genetics and Molecular Research, vol. 13, no. 3, pp. 7912–7918, 2014. [17] O. Waidmann, V. Bihrer, T. Pleli et al., “Serum microRNA-122 levels in different groups of patients with chronic hepatitis B virus infection,” Journal of Viral Hepatitis, vol. 19, no. 2, pp. e58– e65, 2012. [18] K. Arataki, C. N. Hayes, S. Akamatsu et al., “Circulating microRNA-22 correlates with microRNA-122 and represents viral replication and liver injury in patients with chronic hepatitis B,” Journal of Medical Virology, vol. 85, no. 5, pp. 789– 798, 2013. [19] C. Huang, J. M. Zheng, Q. Cheng et al., “Serum microRNA-29 levels correlate with disease progression in patients with chronic hepatitis B virus infection,” Journal of Digestive Diseases, vol. 15, no. 11, pp. 614–621, 2014. [20] F. Yu, J. Yang, J. Ouyang et al., “Serum microRNA-210 levels in different groups of chronic hepatitis B patients,” Clinica Chimica Acta, vol. 450, pp. 203–209, 2015. [21] C.-F. Huang, C.-C. Sun, F. Zhao, Y.-D. Zhang, and D.-J. Li, “miR33a levels in hepatic and serum after chronic HBV-induced fibrosis,” Journal of Gastroenterology, vol. 50, no. 4, pp. 480–490, 2015. [22] Z. Zhang, J. Chen, Y. He et al., “miR-125b inhibits hepatitis B virus expression in vitro through targeting of the SCNN1A gene,” Archives of Virology, vol. 159, no. 12, pp. 3335–3343, 2014. [23] Z. Hou, J. Zhang, Q. Han et al., “Hepatitis B virus inhibits intrinsic RIG-I and RIG-G immune signaling via inducing miR146a,” Scientific Reports, vol. 6, Article ID 26150, 2016.

BioMed Research International [24] Y. Sun, S. Shen, H. Tang et al., “MiR-429 identified by dynamic transcriptome analysis is a new candidate biomarker for colorectal cancer prognosis,” OMICS A Journal of Integrative Biology, vol. 18, no. 1, pp. 54–64, 2014. [25] X. Zhang, E. Zhang, Z. Ma et al., “Modulation of hepatitis B virus replication and hepatocyte differentiation by MicroRNA1,” Hepatology, vol. 53, no. 5, pp. 1476–1485, 2011. [26] Y. Wang, L. Jiang, X. Ji, B. Yang, Y. Zhang, and X.-D. Fu, “Hepatitis B viral RNA directly mediates down-regulation of the tumor suppressor microRNA miR-15a/miR-16-1 in hepatocytes,” Journal of Biological Chemistry, vol. 288, no. 25, pp. 18484–18493, 2013. [27] X. Dai, W. Zhang, H. Zhang et al., “Modulation of HBV replication by microRNA-15b through targeting hepatocyte nuclear factor 1𝛼,” Nucleic Acids Research, vol. 42, no. 10, pp. 6578–6590, 2014. [28] N. Liu, J. Zhang, T. Jiao et al., “Hepatitis B virus inhibits apoptosis of hepatoma cells by sponging the MicroRNA 15a/16 cluster,” Journal of Virology, vol. 87, no. 24, pp. 13370–13378, 2013. [29] E. Petrini, G. P. Caviglia, M. L. Abate, S. Fagoonee, A. Smedile, and R. Pellicano, “microRNA in HBV-related hepatocellular carcinoma: functions and potential cinical applications,” Panminerva Medica, vol. 57, no. 4, pp. 201–209, 2015. [30] E. Connolly, M. Melegari, and P. Landgraf, “Elevated expression of the miR-17-92 polycistron and miR-21 in hepadnavirusassociated hepatocellular carcinoma contributes to the malignant phenotype,” The American Journal of Pathology, vol. 173, no. 3, pp. 856–864, 2008. [31] Y. J. Jung, J.-W. Kim, S. J. Park et al., “c-Myc-mediated overexpression of miR-17-92 suppresses replication of hepatitis B virus in human hepatoma cells,” Journal of Medical Virology, vol. 85, no. 6, pp. 969–978, 2013. [32] W.-H. Liu, S.-H. Yeh, C.-C. Lu et al., “MicroRNA-18a prevents estrogen receptor-alpha expression, promoting proliferation of hepatocellular carcinoma cells,” Gastroenterology, vol. 136, no. 2, pp. 683–693, 2009. [33] P. Damania, B. Sen, S. B. Dar et al., “Hepatitis B virus induces cell proliferation via HBx-induced microRNA-21 in hepatocellular carcinoma by targeting Programmed Cell Death Protein4 (PDCD4) and Phosphatase and Tensin Homologue (PTEN),” PLoS ONE, vol. 9, no. 3, Article ID e91745, 2014. [34] C. Shi and X. Xu, “MicroRNA-22 is down-regulated in hepatitis B virus-related hepatocellular carcinoma,” Biomedicine and Pharmacotherapy, vol. 67, no. 5, pp. 375–380, 2013. [35] Y. Zheng and Z. Xu, “MicroRNA-22 induces endothelial progenitor cell senescence by targeting AKT3,” Cellular Physiology and Biochemistry, vol. 34, no. 5, pp. 1547–1555, 2014. [36] F. Zhao, G. Xu, Y. Zhou et al., “MicroRNA-26b inhibits hepatitis B virus transcription and replication by targeting the host factor CHORDC1 protein,” Journal of Biological Chemistry, vol. 289, no. 50, pp. 35029–35041, 2014. [37] G. Kong, J. Zhang, S. Zhang, C. Shan, L. Ye, and X. Zhang, “Upregulated microRNA-29a by hepatitis B virus X protein enhances hepatoma cell migration by targeting PTEN in cell culture model,” PLoS ONE, vol. 6, no. 5, Article ID e19518, 2011. [38] C.-M. Wang, Y. Wang, C.-G. Fan et al., “MiR-29c targets TNFAIP3, inhibits cell proliferation and induces apoptosis in hepatitis B virus-related hepatocellular carcinoma,” Biochemical and Biophysical Research Communications, vol. 411, no. 3, pp. 586–592, 2011. [39] Y. Wang, C.-M. Wang, Z.-Z. Jiang et al., “MicroRNA-34c targets TGFB-induced factor homeobox 2,represses cell proliferation

BioMed Research International

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

[51]

[52]

[53]

[54]

and induces apoptosis in hepatitis B virus-related hepatocellular carcinoma,” Oncology Letters, vol. 10, no. 5, pp. 3095–3102, 2015. M. Ohno, M. Otsuka, T. Kishikawa et al., “Specific delivery of microRNA93 into HBV-replicating hepatocytes downregulates protein expression of liver cancer susceptible gene MICA,” Oncotarget, vol. 5, no. 14, pp. 5581–5590, 2014. J. Zhang, H. Jin, H. Liu et al., “MiRNA-99a directly regulates AGO2 through translational repression in hepatocellular carcinoma,” Oncogenesis, vol. 3, no. 4, article no. e97, 2014. Y. Wang and H. Tian, “miR-101 suppresses HBV replication and expression by targeting FOXO1 in hepatoma carcinoma cell lines,” Biochemical and Biophysical Research Communications, vol. 487, no. 1, pp. 167–172, 2017. L. Xu, S. Beckebaum, S. Iacob et al., “MicroRNA-101 inhibits human hepatocellular carcinoma progression through EZH2 downregulation and increased cytostatic drug sensitivity,” Journal of Hepatology, vol. 60, no. 3, pp. 590–598, 2014. X. Wei, T. Xiang, G. Ren et al., “MiR-101 is down-regulated by the hepatitis B virus x protein and induces aberrant DNA methylation by targeting DNA methyltransferase 3A,” Cellular Signalling, vol. 25, no. 2, pp. 439–446, 2013. W. Deng and M. Lu, “The role of microRNAs in hepatocyte metabolism and hepatitis B virus replication,” Virologica Sinica, vol. 31, no. 6, pp. 472–479, 2016. Y. Naito, Y. Tanaka, and T. Ochiya, “MicroRNAs and hepatitis B,” Advances in Experimental Medicine and Biology, vol. 888, pp. 389–399, 2015. J. Lamontagne, L. F. Steel, and M. J. Bouchard, “Hepatitis B virus and microRNAs: Complex interactions affecting hepatitis B virus replication and hepatitis B virusassociated diseases,” World Journal of Gastroenterology, vol. 21, no. 24, pp. 7375–7399, 2015. N. Mosca, F. Castiello, N. Coppola et al., “Functional interplay between hepatitis B virus X protein and human miR-125a in HBV infection,” Biochemical and Biophysical Research Communications, vol. 449, no. 1, pp. 141–145, 2014. M. Ninomiya, Y. Kondo, O. Kimura et al., “The expression of MIR-125b-5p is increased in the serum of patients with chronic hepatitis B infection and inhibits the detection of hepatitis B virus surface antigen,” Journal of Viral Hepatitis, vol. 23, no. 5, pp. 330–339, 2016. W. Deng, X. Zhang, Z. Ma, Y. Lin, and M. Lu, “MicroRNA-125b5p mediates post-transcriptional regulation of hepatitis B virus replication via the LIN28B/let-7 axis,” RNA Biology, pp. 1–10, 2017. J.-Y. Huang, S.-F. Chou, J.-W. Lee et al., “MicroRNA-130a can inhibit hepatitis B virus replication via targeting PGC1𝛼 and PPAR𝛾,” RNA, vol. 21, no. 3, pp. 385–400, 2015. F. Gao, X. Sun, L. Wang, S. Tang, and C. Yan, “Downregulation of MicroRNA-145 caused by Hepatitis B Virus X protein promotes expression of CUL5 and contributes to pathogenesis of Hepatitis B virus-associated hepatocellular carcinoma,” Cellular Physiology and Biochemistry, vol. 37, no. 4, pp. 1547–1559, 2015. J.-F. Li, X.-P. Dai, W. Zhang et al., “Upregulation of microRNA146a by hepatitis B virus X protein contributes to hepatitis development by downregulating complement factor H,” mBio, vol. 6, no. 2, Article ID e02459-14, 2015. Y. Cao, J. Chen, D. Wang et al., “Upregulated in Hepatitis B virus-associated hepatocellular carcinoma cells, miR-331-3p promotes proliferation of hepatocellular carcinoma cells by targeting ING5,” Oncotarget, vol. 6, no. 35, pp. 38093–38106, 2015.

17 [55] H. Gao and C. Liu, “miR-429 represses cell proliferation and induces apoptosis in HBV-related HCC,” Biomedicine and Pharmacotherapy, vol. 68, no. 8, pp. 943–949, 2014. [56] G. Zhang, N. Li, Z. Li et al., “MicroRNA-4717 differentially interacts with its polymorphic target in the PD1 3’ untranslated region: A mechanism for regulating PD-1 expression and function in HBV-associated liver diseases,” Oncotarget, vol. 6, no. 22, pp. 18933–18944, 2015. [57] J.-G. Joung, K.-B. Hwang, J.-W. Nam, S.-J. Kim, and B.-T. Zhang, “Discovery of microRNA-mRNA modules via populationbased probabilistic learning,” Bioinformatics, vol. 23, no. 9, pp. 1141–1147, 2007. [58] K. D. Taganov, M. P. Boldin, K. Chang, and D. Baltimore, “NF𝜅B-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 33, pp. 12481–12486, 2006. [59] G. Curtale, F. Citarella, C. Carissimi et al., “An emerging player in the adaptive immune response: MicroRNA-146a is a modulator of IL-2 expression and activation-induced cell death in T lymphocytes,” Blood, vol. 115, no. 2, pp. 265–273, 2010. [60] G.-L. Zhang, Y.-X. Li, S.-Q. Zheng, M. Liu, X. Li, and H. Tang, “Suppression of hepatitis B virus replication by microRNA199a-3p and microRNA-210,” Antiviral Research, vol. 88, no. 2, pp. 169–175, 2010. [61] P. Wang, C. Zhuang, D. Huang, and K. Xu, “Downregulation of miR-377 contributes to IRX3 deregulation in hepatocellular carcinoma,” Oncology Reports, vol. 36, no. 1, pp. 247–252, 2016. [62] G. Szabo and S. Bala, “MicroRNAs in liver disease,” Nature Reviews Gastroenterology & Hepatology, vol. 10, no. 11, pp. 542– 552, 2013. [63] L. Gramantieri, F. Fornari, E. Callegari et al., “MicroRNA involvement in hepatocellular carcinoma,” Journal of Cellular and Molecular Medicine, vol. 12, no. 6, pp. 2189–2204, 2008.

Expression Profiling of Cellular MicroRNA in Asymptomatic HBsAg Carriers and Chronic Hepatitis B Patients.

MicroRNAs (miRNAs) may serve as potential molecular markers to predict liver injury resulting from chronic hepatitis B (CHB). In the present study, we...
2MB Sizes 0 Downloads 15 Views