30
1 IFN-α-induced miR-122 inhibition negatively affects the anti-HBV 1 efficiency of IFN-α 2 3 Junli Hao a , Wensong Jin a , Xinghui Li a , Saifeng Wang a , Xiaojun Zhang a , Hongxia 4 Fan a , Changfei Li a , Lizhao Chen a , Bin Gao a , Guangze Liu b *, Songdong Meng a * 5 6 a CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of 7 Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101. 8 b Transgenic Engineering Research Laboratory, Infectious Disease Center, 458th 9 Hospital, Guangzhou, China. 10 11 *Corresponding authors. Tel: +86-10-64807350; Fax: +86-10-64807381. 12 E-mail address: [email protected] (G. Liu); [email protected] (S. Meng); 13 14 Running title: miR-122 sponge limits anti-HBV effect of IFN-α 15 16 ABSTRACT 17 Interferon (IFN)-α based therapy can effectively treat chronic hepatitis B virus 18 (HBV) infection which causes life-threatening complications. Responses to IFN-α 19 therapy vary greatly in chronic hepatitis B (CHB) patients but underlying mechanisms 20 are almost unknown. In this study, we found that IFN-α treatment induced marked 21 decrease of miR-122 expression in hepatocytes. We next showed that IFN-α-induced 22 miR-122 down-regulation was only partly due to transcriptional suppression. One 23 IFN-stimulated gene (ISG) NT5C3 which was identified as a miR-122 target, 24 efficiently inhibited miR-122 by binding and sequestering miR-122 with its mRNA 25 3’-UTR, indicating that ISG is involved in IFN-α-mediated miR-122 suppression. 26 Notably, the inhibitory effect of IFN-α on miR-122 was completely abolished by 27 blocking IFN-α-induced up-regulation of NT5C3 mRNA expression by RNAi. 28 Meanwhile, we observed that miR-122 dramatically inhibited HBV expression and 29 replication. Finally, we showed that IFN-α-mediated HBV inhibitory effects could be 30 Copyright © 2012, American Society for Microbiology. All Rights Reserved. J. Virol. doi:10.1128/JVI.01710-12 JVI Accepts, published online ahead of print on 10 October 2012

Inhibition of Alpha Interferon (IFN- )-Induced MicroRNA-122 Negatively Affects the Anti-Hepatitis B Virus Efficiency of IFN- 

Embed Size (px)

Citation preview

1

IFN-α-induced miR-122 inhibition negatively affects the anti-HBV 1

efficiency of IFN-α 2

3

Junli Haoa, Wensong Jina, Xinghui Lia, Saifeng Wanga, Xiaojun Zhanga , Hongxia 4

Fana, Changfei Lia, Lizhao Chena, Bin Gaoa, Guangze Liu b*, Songdong Menga* 5

6

a CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of 7

Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101. 8

b Transgenic Engineering Research Laboratory, Infectious Disease Center, 458th 9

Hospital, Guangzhou, China. 10

11

*Corresponding authors. Tel: +86-10-64807350; Fax: +86-10-64807381. 12

E-mail address: [email protected] (G. Liu); [email protected] (S. Meng); 13

14

Running title: miR-122 sponge limits anti-HBV effect of IFN-α 15

16

ABSTRACT 17

Interferon (IFN)-α based therapy can effectively treat chronic hepatitis B virus 18

(HBV) infection which causes life-threatening complications. Responses to IFN-α 19

therapy vary greatly in chronic hepatitis B (CHB) patients but underlying mechanisms 20

are almost unknown. In this study, we found that IFN-α treatment induced marked 21

decrease of miR-122 expression in hepatocytes. We next showed that IFN-α-induced 22

miR-122 down-regulation was only partly due to transcriptional suppression. One 23

IFN-stimulated gene (ISG) NT5C3 which was identified as a miR-122 target, 24

efficiently inhibited miR-122 by binding and sequestering miR-122 with its mRNA 25

3’-UTR, indicating that ISG is involved in IFN-α-mediated miR-122 suppression. 26

Notably, the inhibitory effect of IFN-α on miR-122 was completely abolished by 27

blocking IFN-α-induced up-regulation of NT5C3 mRNA expression by RNAi. 28

Meanwhile, we observed that miR-122 dramatically inhibited HBV expression and 29

replication. Finally, we showed that IFN-α-mediated HBV inhibitory effects could be 30

Copyright © 2012, American Society for Microbiology. All Rights Reserved.J. Virol. doi:10.1128/JVI.01710-12 JVI Accepts, published online ahead of print on 10 October 2012

2

significantly enhanced when blocking IFN-α-induced down-regulation of miR-122. 31

We therefore conclude that IFN-α-induced inhibition of miR-122 may negatively 32

affect the anti-HBV function of IFN-α. These data provide valuable insight for better 33

understanding of the anti-viral mechanism of IFN-α and raise further potential interest 34

in enhancing its anti-HBV efficacy. 35

36

Around 400 million people worldwide are infected with hepatitis B virus (HBV). 37

Chronic hepatitis B (CHB), which is triggered by HBV infection, poses a huge health 38

burden on the global community as it is correlated with a significant increased risk for 39

the development of cirrhosis, liver failure and hepatocellular carcinoma (HCC) (24). 40

Currently the treatment of CHB mainly consists of pegylated interferon (IFN)-α and 41

nucleoside or nucleotide analogs (e.g. lamivudine, adefovir, and entecavir). IFN-α 42

was the first drug licensed to treat HBV infection. As an important first-line treatment 43

option, pegylated IFN-α can effectively treat CHB in 25-40% of patients as 44

monotherapy, and greater sustained virological response (SVR) and hepatitis B e 45

antigen (HBeAg) seroconversion rates in HBeAg-positive patients were observed with 46

addition of nucleos(t)ide analogue therapies (29, 37). In fact, treatment with pegylated 47

IFN results in the highest rate of off-treatment sustained response among currently 48

available drugs (5). Moreover, responses to IFN-based therapy are accompanied by 49

the potential for hepatitis B surface antigen (HBsAg) loss or seroconversion, and early 50

serum HBsAg loss has been recently reported to have predictive values of SVR to 51

IFN in both HBeAg positive and negative CHB patients (23, 35, 47). 52

As a member of type I interferons, IFN-α can initiate activation of Jak/STAT and 53

NF-κB signaling, which induces hundreds of IFN-stimulated genes (ISGs) and may 54

play an important role in IFN-mediated anti-HBV activity. Both in vitro and in vivo 55

studies showed that besides a stimulating effect on cytotoxic T lymphocytes and 56

natural killer cell function, IFN-based therapy (IFN-α-2b and pegylated IFN-α-2a or 57

-2b) also has a direct anti-viral effect by preventing the formation or accelerating 58

decay of viral capsids, and/or inducing anti-viral ISGs that inhibit HBV expression 59

and replication (50, 21, 30, 44, 43, 51). Inhibition of IFN-α signaling by HBV has 60

3

been suggested to antagonize the IFN response (20). Nevertheless, these studies also 61

strongly suggest that there is in principle significant potential to modulate the 62

effectiveness of IFN-mediated anti-HBV activities. Moreover, the anti-viral activity of 63

ISGs remains elusive and still awaits further investigation (39). Responses to IFN-α 64

therapy vary greatly in CHB patients but underlying mechanisms are almost unknown 65

(23, 35, 5). Notably, IFNα/β was recently found to suppress HBV replication when 66

viral load is high whereas enhance HBV replication when viral load is low in HBV 67

transgenic mice, indicating its dual function for HBV (38). Taken together, the precise 68

mechanism of action of IFN-α has not been fully understood. 69

MicroRNAs (miRNAs) are a class of small RNAs of approximately 22 70

nucleotides in length, which interact with complementary target sites usually in the 71

3'-untranslated region (UTR) of target mRNAs and induce their translational 72

repression, deadenylation and degradation. MicroRNA-122 (miR-122), a mammalian 73

liver-specific microRNA, is highly expressed in the liver, which constitutes 70% of 74

the total miRNA population. There is growing literature on biological functions of 75

miR-122 in the control of hepatocyte growth and neoplastic transformation (10, 49, 4, 76

46), regulation of lipid metabolism (22), liver development (45) and modulation of 77

HBV and hepatitis C virus (HCV) replication (17, 19, 25, 28, 8). 78

Previous studies showed that IFN-β treatment leads to a significant decrease in the 79

expression of miR-122 both in vitro and in vivo (27, 31), but its regulatory 80

mechanisms are totally unknown. In addition, our recent study showed that miR-122 81

inhibits HBV replication and transcription through cyclin G1-modulated P53 activity 82

(42). Given the importance of IFN-α-based therapy against HBV and the essential 83

roles of miR-122 in hepatic function, it is worthwhile to assess the regulation of 84

miR-122 in response to IFN-α and its impact on HBV replication. We identified at 85

least one IFN-α induced ISG as a miR-122 target gene, which may act as a sponge to 86

bind and sequester endogenous miR-122. The results could shed light on the 87

mechanism of action of IFN-α on HBV replication. 88

89

Materials and methods 90

4

Reagents and antibodies. The chemically synthesized miR-122 inhibitor and 91

non-specific control, the miR-122 mimic and non-specific control were purchased 92

from RiboBio Co., Ltd. (Guangzhou, China). The sequence of miR-122 mimic was 93

obtained from the miRBase Database (http://www.mirbase.org) 5’-3’: UGGAGUGU 94

GACAAUGGUGUUUG and the sequence of miR-122 inhibitor is completely 95

complementary to miR-122. The siRNAs of NT5C3 and p53 were synthesized from 96

RiboBio Co., Ltd. (Guangzhou, China). The sense and antisense NT5C3 siRNA were 97

5’-3’: CCAACAUGUCAUAAUAUCAdTdT (sense), 5’-3’: UGAUAUUAUGACAU 98

GUUGGdTdT (antisense), and the sense and antisense p53 siRNA were 5’-3’: 99

GACUCCAGUGGUAAUCUACdTdT (sense), 5’-3’: GUAGAUUACCACUGGAGU 100

CdTdT (antisense). The following reagents and antibodies were obtained as indicated: 101

IFN-α (Recombinant Human Interferon α-2b injection, Anke Biotechnology, Anhui, 102

China); Deoxyribonuelease�(D8071-25, Solarbio Science & technology Co., Ltd, 103

Beijing, China); Proteinase K (D9033, Takara BioInc., Shiga, Japan); DNase I 104

(RNase-free) (Invitrogen); SuperscriptTM RT reagent kit (DRR037A, Takara BioInc., 105

Shiga, Japan); the rabbit anti-human cyclin G1 (clone F-5, Santa Cruz Biotechnology); 106

the mouse anti-p53 monoclonal antibody (clone 1C12, Cell Signaling Technology); 107

the mouse anti-human actin and the horseradish peroxidase-conjugated secondary 108

antibodies (Zhongshan Goldenbridge Biotechnology, China); and the ECL-Plus 109

chemiluminescence system (Applygen Technologies, Beijing, China). 110

Cell culture and constructs. Two human hepatoma cell lines (Huh-7 and HepG2) 111

were obtained from the ATCC (Manassas, VA, USA). For transfection studies, Huh-7 112

and HepG2 cells were washed twice with Opti-MEM (Invitrogen), and miR-122 113

inhibitor or mimic was transfected using Lipofectamine 2000 (Invitrogen). HBV 114

replication plasmid pHBV, containing 1.3 copies of the HBV genome (D genotype) 115

(36), was maintained in the lab. HBV-luciferase plasmid (pHBV-Luc), a HBV 116

expressing construct containing a luciferase ORF under the HBV enhancers and core 117

promoter, is a gift kindly provided by Dr. Yosef Shaul (The Weizmann Institute of 118

Science, Israel) (34). The construct pGL-122 with a luciferase reporter gene under 119

miR-122 promoter was provided by Dr. Shi-Mei Zhuang (Sun Yat-sen University, 120

5

China) (49). 121

Southern blot and Northern blot analysis. Core-particle related HBV DNA was 122

isolated as described (26). The HBV DNA was analyzed by Southern blot 123

hybridization using AP-labeled HBV DNA as the probe (42). For miRNA Northern 124

blot, the probes were labeled at their 5′-end using polynucleotide kinase (New 125

England Biolabs) and [γ-32P]ATP. The probe sequence of miR-122 was completely 126

complementary to miR-122 (5’-3’: CAAACACCATTGTCACACTCCA), and the 127

sequence of the U6 snRNA probe was 5’-3’: GCAGGGGCCATGCTAATCTTCTCTG 128

TATCG. Total RNA was isolated using the Trizol Reagent (Invitrogen, USA). 30μg of 129

total RNA for each lane was denatured and separated on 17% polyacrylamide gels, 130

transferred to Hybond-N Plus membrane (GE Healthcare) by semi-dry blotting, and 131

hybridized with the labeled probes overnight. Blots were washed and the 32P blot was 132

exposed to film. 133

Immunoblot analysis. An equal amount of protein from each cell lysate was 134

separated in a 15% SDS–polyacrylamide gel and probed with an anti-p53 or 135

anti-cyclinG1 and an anti-human actin antibody. The bound antibodies were 136

visualized using an ECL detection kit with appropriate HRP-conjugated secondary 137

antibodies (Santa Cruz Biotech.). 138

Luciferase Reporter Assays. Dual luciferase reporter assays were performed 139

following the manufacturer’s protocol (Promega). The 3’-UTR of NT5C3 was cloned 140

into downstream of the firefly luciferase gene in the pGL3 plasmid. Huh-7 cells were 141

transfected with a firefly luciferase reporter plasmid carrying either the wild-type 142

(NT5C3-wt) or mutant (NT5C3-mut) 3’-UTR of NT5C3. At 48 hr post-transfection, 143

firefly luciferase and Renilla luciferase activities were measured, and the luciferase 144

activity was normalized to Renilla luciferase. 145

RNA, DNA extraction and real-time PCR. Total RNA was extracted from treated 146

cells using the Trizol Reagent (Invitrogen). RNA extracts were treated with 147

RNase-free DNAse I (Invitrogen), as recommended by the manufacturer. 500 ng of 148

cellular RNA was reverse transcribed with 0.5μl each of oligo dT and Random 6 mers 149

primers by using the SuperscriptTM RT reagent kit (Takara), and samples processed 150

6

without reverse transcriptase were used as negative controls to examine residual DNA 151

contamination (40). Specific primers used to detect HBV pgRNA and total mRNAs 152

by real time PCR were as follows: pgRNA: forward primer, 5’-TCTTGCCTTACTTT 153

TGGAAG-3’, reverse primer, 5’-AGTTCTTCTTCTAGGGGACC-3’, and total 154

mRNAs designed on HBx region: forward primer, 5′-ACGTCCTTTGTTTACGTCCC 155

GT-3′, reverse primer, 5′-CCCAACTCCTCCCAGTCCTTAA-3′ (1). GAPDH was 156

used as an internal standard for the quantification of the PCR. The primers used to 157

detect NT5C3 and GAPDH have been described previously (33, 48). 158

Cells were washed three times with PBS to remove the input plasmid DNA after 159

pHBV transfection. After 24 or 48 hr culture, the cell supernatants were harvested and 160

digested with 100 UI/ml DNAaseⅠat 37°C for at least 3 hours to remove the input 161

plasmid. The supernatants were then digested with 1% sodium dodecyl sulfate, 0.5 162

mg/ml proteinase K at 55°C for 2 hours to release viral DNA in the capsids. Viral 163

DNA copy numbers were detected by real time PCR. The primes designed on HBx 164

region were as follows: forward primer 5′-ACGTCCTTTGTTTACGTCCCGT-3′ (nt 165

1414-1435), reverse primer 5′-CCCAACTCCTCCCAGTCCTTAA-3′ (nt 1744-1723) 166

(1, 41). The supernatants without proteinase digestion were used as the negative 167

control. No HBV DNA in the control supernatants were detected by real-time PCR, 168

indicating that the input plasmid was totally removed. 169

Real-time fluorescence quantitative PCR was performed using the SYBR Green 170

Premix Reagent (Takara BioInc., Shiga, Japan). 2 μL of cDNA or DNA was used as 171

input template for each real-time PCR assay. Each experiment was performed in 172

triplicate. PCR cycling conditions were two steps as follows: 30 s at 95°C, followed 173

by 40 cycles of 95°C for 15 s and 60°C for 45 s. Melt-curve analysis was used to 174

estimate the specificity of primers. 175

Preparation of standard HBV DNA. PCR product for HBx (nt 1374-1838) was 176

cloned into pcDNA3.1 (Invitrogen). The plasmid was quantified using a NanoDrop at 177

OD 260 nm (1). For a standardized evaluation, the number of HBV DNA copies/ml in 178

the cell supernatant was calculated from a standard curve with 10-fold series dilution 179

7

of the plasmid pcDNA3.1-HBx ranging from 103 to 109 HBV copies/ml. 180

TaqMan miRNA analysis. Real-time PCR analysis for miR-122 was performed 181

using the TaqMan miRNA analysis Kit (Applied Biosystems, Foster City, CA, USA). 182

A U6 endogenous control was used for normalization. Pri-miR-122 was detected 183

using High Capacity cDNA Reverse Transcription Kit (Applied Biosystems) and 184

TaqMan pri-miRNA Assay. Actin was used as an internal standard gene. 185

Detection of HBsAg and HBeAg by enzymed-linked immunosorbent assay 186

(ELISA). ELISA tests for detection of HBsAg and HBeAg were performed as 187

described previously (28). 188

Statistical analysis. Each experiment was performed in triplicate, and all 189

experiments were repeated at least two times. The results are presented as the means ± 190

standard deviation. Student’s t-test was used for comparison between groups. A p 191

value <0.05 was considered a significant difference. 192

193

Results 194

IFN-α induces down-regulation of miR-122 and suppresses HBV replication. 195

In this study, Huh-7 cells were selected for testing whether miR-122 was subject to 196

regulation by IFN-α because this is the only human hepatoma cell line with 197

constitutive expression of miR-122. We first analyzed miR-122 expression in 198

response to IFN-α in Huh-7 cells by real-time PCR. As shown in Fig.1A, 199

down-regulation of miR-122 followed a dose-response curve between 1 and 1000 200

U/ml of IFN-α at 4 hr after treatment. Treatment with 1000 U/ml of IFN-α caused 201

about 60% reduction of miR-122. Time course analysis showed a decrease of 202

miR-122 levels with maximum between 4 and 8 hr, which almost reached to normal 203

level at 48 hr (Fig. 1B). To determine if the restoration of miR-122 level was due to 204

IFN-α degradation, we added 1000 U/ml of IFN-α to the cell culture for every 12 hr. 205

As seen in Fig. 1C and D, persistent decreases of miR-122 were observed by real time 206

PCR and Northern blot assays. In the mean time, Huh-7 cells transfected with the 207

HBV replication plasmid pHBV were treated with 1000 U/ml of IFN-α. Significant 208

decreases of HBsAg and HBeAg expression were detected in Huh-7 cells at 24, 48 209

8

and 72 hr after treatment with IFN-α (Fig. 1E), and significant reduction of 210

HBV-DNA levels in the cell supernatant was observed at all time points (Fig. 1F). We 211

further quantified HBV replication by Southern blot analysis (Fig. 1G). Significant 212

reductions in HBV replicative intermediates were observed in IFN-α-treated cells. 213

Consistently, treatment with IFN-α significantly decreased HBV pgRNA and total 214

mRNA transcripts (Fig. 1H), suggesting that IFN-α affects HBV replication at least 215

partly at the transcriptional level. 216

IFN-α-induced ISG with a complementary binding site to miR-122 sequesters 217

endogenous miR-122. Next, we explored the mechanism of IFN-α-induced miR-122 218

down-regulation. To evaluate the role of IFN-α in regulation of miR-122 transcription, 219

we performed a luciferase reporter assay using the construct pGL-122 with a 220

luciferase reporter gene under miR-122 promoter (49). IFN-α caused moderate 221

reductions (about 20-30%) in the promoter activity at 12 and 24 hr after treatment 222

(Fig. 2A). As can be seen in Fig. 1B, maximum decrease of miR-122 level occurred 223

around 4 hr post-treatment, whereas there was no significant change in miR-122 224

transcriptional activity at this time period, indicating that IFN-α affects miR-122 level 225

not mainly through transcriptional modulation. Real-time PCR assay also revealed no 226

obvious change in miR-122 primary transcript (pri-miR-122) levels between 4 and 8 227

hr after IFN-α treatment (Fig. 2B), validating that IFN-α does not influence miR-122 228

transcription. 229

Considering the quick decrease of miR-122 and hundreds of induced ISGs by 230

IFN-α treatment, we hypothesized that IFN-α-induced mRNAs which harbor miR-122 231

complementary binding sites may sequester and deplete endogenous miR-122. To 232

search for the potential “sponge” mRNAs to miR-122, we focused primarily on the 233

verified genes which are significantly induced by IFN-α (12). Among 107 genes 234

identified as significantly upregulated at 6 hr after IFN-α treatment, the 3’-UTRs of 235

cytoplasmic 5'-nucleotidase-III (NT5C3) and Interleukin-1 receptor antagonist 236

(IL-1RN) were identified as predicted targets of miR-122 by TargetScan 237

(http://www.targetscan.org). We focused on NT5C3 because transfection with its 238

3’-UTR in Huh-7 cells led to more decreased miR-122 levels compared with IL-1RN 239

9

3’-UTR. A putative miR-122 complementary region in the NT5C3 sequence (position 240

183-189 of NT5C3 3’-UTR) is listed in Fig. 3A. Obviously, miR-122 reduced the 241

activity of firefly luciferase with the wild-type but not mutant 3’-UTR of NT5C3. As 242

shown in Fig. 3B, compared with the mutant 3’-UTR, ectopic overexpression of the 243

wild-type 3’-UTR of NT5C3 by transfection with pGL-NT5C3wt vector in Huh-7 244

cells led to decreased miR-122 levels by 82% and 43% respectively at 24 and 48 hr 245

post-transfection by real-time PCR analysis (both P<0.01). Decrease of miR-122 was 246

confirmed by Northern blot (Fig. 3C). In addition, inhibition of miR-122 by the 247

3’-UTR of NT5C3 was further confirmed by Western blot analysis of expression of a 248

verified miR-122 target cyclin G1 (11). Transfection of miR-122 mimic led to a 249

decreased expression of cyclin G1, whereas the depletion of endogenous miR-122 by 250

its inhibitor resulted in an increase of cyclin G1 (Fig. 3D). As can be seen in Fig. 3E, 251

the wild-type but not the mutant 3’-UTR transfected Huh-7 cells showed an increase 252

of cyclin G1 expression, indicating that the 3’-UTR of NT5C3 rescued the expression 253

of the miR-122 target cyclin G1 by inhibition of miR-122. Moreover, time course 254

analysis showed dramatic increase (about 2.5-fold) of NT5C3 mRNA levels with 255

maximum between 4 and 12 hr after treatment with IFN-α (Fig. 3F). As demonstrated 256

in Fig. 1B, decrease of miR-122 levels was synchronized with the increase of NT5C3 257

mRNA levels. 258

To further demonstrate that IFN-α-induced NT5C3 mRNA sequesters endogenous 259

miR-122, Huh-7 cells were treated with NT5C3 siRNA. Significant increase of 260

miR-122 was observed by real-time PCR and Northern blot analyses (Fig. 4A). Next, 261

Huh-7 cells were treated with IFN-α and NT5C3 siRNA, as indicated in Fig. 4B. 262

Analysis of NT5C3 expression by real-time PCR revealed approximately equal 263

amount of mRNA levels between IFN-α plus NT5C3 RNAi treated cells and untreated 264

cells, suggesting that up-regulation of NT5C3 by IFN-α could be attenuated by 265

transfection of NT5C3 siRNA. As expected, IFN-α treatment caused a decrease of 266

miR-122, whereas knock-down of NT5C3 led to an increased miR-122 level. Notably, 267

the inhibitory effect of IFN-α on miR-122 was completely abolished by blocking 268

IFN-α-induced up-regulation of NT5C3 mRNA expression using RNAi (P>0.05). 269

10

Taken together, these results suggest that the rapid decline of miR-122 induced by 270

IFN-α occurs via up-regulation of NT5C3 mRNA expression and the subsequent 271

saturation and depletion of miR-122 by binding to the complementary binding site 272

located in the 3’-UTR of NT5C3. 273

Inhibition of miR-122 by IFN-α can compromise the anti-HBV activity of 274

IFN-α. To investigate the potential effect of IFN-α-induced miR-122 down-regulation 275

on HBV replication, Huh-7 cells were treated with 25 nM of chemically synthesized 276

miR-122 inhibitor or 25 nM miR-152 or miR-642 as negative controls or a 277

randomized oligonucleotide control as mock 24 hr before transfection with pHBV. 278

Compared with mock-treated cells, significant increases of HBsAg and HBeAg 279

expression were detected in Huh-7 cells after treatment with miR-122 inhibitor at 48 280

hr after transfection (both P<0.01) (Fig. 5A). Treatment with miR-122 inhibitor led to 281

an increase of HBV DNA level by about 4-fold (Fig. 5B). We chose to use 25 nM of 282

miR-122 inhibitor for the experiment because this concentration caused about 50-60% 283

reduction of miR-122 (data not shown), which was the similar amount of 284

down-regulation induced by IFN-α (Fig. 1A and B). 285

Conversely, overexpression of miR-122 was performed in HepG2 cells as the cells 286

express very low level of miR-122. HepG2 cells were co-transfected with pHBV 287

together with 50 nM of chemically synthesized miR-122 mimic or 50nM miR-152 or 288

miR-642 as negative controls or a randomized oligonucleotide control as mock. A 289

significant decrease of HBsAg and HBeAg expression was observed 48 hr after 290

miR-122 transfection (both P<0.01) (Fig. 5C). Treatment with miR-122 mimic led to 291

decreases of HBV total mRNA level by about 50% (Fig. 5D), and DNA level by 292

about 64% (Fig. 5E), and a dramatic reduction in HBV replication was observed by 293

Southern blot analysis (Fig. 5F). The results were consistent with the finding in Huh-7 294

cells that miR-122 inhibits HBV replication. 295

The miR-122 level in Huh-7 cells was decreased by about 50-60% under IFN-α 296

treatment, which was similar to the reduction induced by miR-122 inhibitor. The 297

decrease of miR-122 by its inhibitor resulted in significant increase of HBV 298

replication. We therefore conclude that inhibition of miR-122 by IFN-α negatively 299

11

impacts the IFN-α-mediated inhibition of HBV. 300

To rule out the possibility that competition for RISC complexes may interfere with 301

test results, we also used miR-152 and miR-642 as negative controls in the NT5C3 302

luciferase reporter assay (Fig. 3A) and HBV expression and replication assay (Fig. 5), 303

and no obvious differences were observed among the random control oligonucleotides, 304

miR-152 and miR-642, indicating the miR-122-specific effects in these assays. 305

To further demonstrate that IFN-α-mediated regulation of miR-122 through NT5C3 306

plays a negative role in the HBV inhibitory function of IFN-α, Huh-7 cells were 307

transfected with pGL-NT5C3wt vector to mimic IFN-α-induced up-regulation of 308

NT5C3 expression. As seen in Fig. 6A, overexpression of the 3’-UTR of NT5C3 309

resulted in pronounced increase of HBV replication compared with transfection with 310

pGL-NT5C3mut vector carrying mutant 3’-UTR of NT5C3 (P<0.01). Furthermore, 311

pHBV transfected Huh-7 cells were treated with 500 U/ml of IFN-α and 25 nM of 312

miR-122 mimic, as indicated in Fig. 6B. As expected, IFN-α treatment and miR-122 313

mimic caused a significant decrease of HBV replication levels. Notably, the inhibitory 314

effect was greatly enhanced by blocking IFN-α-induced down-regulation of miR-122 315

by mimic transfection (P<0.05 or 0.01). In addition, pHBV-transfected Huh-7 cells 316

were treated with 1000 U/ml of IFN-α and 50 nM of miR-122 inhibitor, as indicated 317

in Fig. 6C. IFN-α treatment caused 31.2% reduction of HBV DNA level in 318

mock-treated Huh-7 cells, while it caused 46.7% reduction of HBV DNA level in 319

miR-122 inhibitor-treated cells, indicating the anti-HBV efficiency of IFN-α could be 320

increased under miR-122 depletion. The data support the notion that down-regulation 321

of miR-122 by IFN-α plays a negative role in the anti-HBV activity of IFN-α. 322

Finally, as our previous study showed that miR-122 inhibits HBV replication 323

through transcription repression by cyclin G1-mediated p53 activity (42), we 324

determined to investigate if the mutant p53 (Tyr220Cys) in Huh-7 cells could target 325

HBV enhancers and regulate viral transcription and replication. Huh-7 was 326

cotransfected with p53-specific siRNA and an HBV-luciferase plasmid (pHBV-Luc), 327

a HBV expressing construct containing a luciferase ORF under the HBV enhancer I/II 328

and core promoter. Transfection of p53 RNAi led to decrease of p53 in both mRNA 329

12

(Fig. 7A) and protein levels (Fig. 7B). Depletion of p53 in Huh-7 cells caused a 330

significant increase in the HBV promoter activity (P<0.01), indicating the mutant p53 331

could regulate HBV transcription. Moreover, p53 RNAi led to increased secretion of 332

HBsAg and HBeAg (Fig. 7D), and a clear increase (more than 1-fold) of viral DNA 333

level in the supernatant (Fig. 7E). The result is consistent to the finding of a previous 334

study showing that the mutant p53 with an increased protein level and prolonged 335

half-life still maintains the transcription activity in Huh-7 cells (13). 336

Together, these data indicate that down-regulation of miR-122 by IFN-α plays a 337

negative role in the anti-HBV activity of IFN-α. 338

339

Discussion 340

IFN-α has been extensively used as one of the major standard treatments for CHB 341

for more than 2 decades. However, its anti-viral activity is limited and the mechanism 342

against HBV awaits further investigation (39, 44, 38, 23, 35). Here, we demonstrated 343

that IFN-α inhibited miR-122 by transcriptional suppression, as well as by inducing at 344

least one ISG NT5C3 which mRNA acted as a sponge to bind and sequester 345

endogenous miR-122. Moreover, our finding revealed that IFN-α-induced inhibition 346

of the most abundant liver-specific microRNA miR-122 may negatively affect the 347

anti-HBV efficiency of IFN-α. This conclusion was further supported by our 348

observations that the inhibitory effect of IFN-α on HBV can be significantly enhanced 349

by blocking IFN-α-induced miR-122 down-regulation (Fig. 6B), and that transfection 350

with the NT5C3 3’-UTR expression vector, which mimics IFN-α-induced NT5C3 351

expression, led to increased HBV replication (Fig. 6A). In addition, the results suggest 352

that IFN-α and miR-122 may both inhibit HBV replication at transcriptional level (see 353

Fig. 1H and Fig. 5D). Our current work may therefore provide further dissection of 354

the anti-viral mechanism of IFN-α, and may help to design a more efficient 355

combination anti-HBV therapy for this first-line drug. 356

The miRNA sponge method by delivering the sponge transgene with multiple 357

target sites of complementary to a miRNA of interest, has been widely used for 358

miRNA loss-of-function studies (7, 9, 18). The phenomenon of endogenous miRNA 359

13

saturation or sponge by the target mRNAs was noted in previous studies (15, 6), but 360

the precise mechanism has not been formally documented. In this study we first 361

systematically screened more than one hundred ISGs induced by IFN-α to explore the 362

potential targets of miR-122, and identified at least one IFN-induced gene, NT5C3, 363

which harbors a putative miR-122 complementary region in its mRNA 3’-UTR. We 364

then checked whether NT5C3 mRNA 3’-UTR could inhibit miR-122. The results 365

showed that overexpression of the 3’-UTR significantly reduced miR-122 levels and 366

its activity. Moreover, we observed about 2.5-fold induction of NT5C3 mRNA in 367

IFN-α-treated cells compared with untreated cells, and the decrease of miR-122 was 368

perfectly synchronized with the increase of NT5C3 mRNA levels at 4 and 8 hr 369

post-treatment, whereas miR-122 transcription levels did not show any apparent 370

changes at these time points. Importantly, miR-122 inhibitory effect of IFN-α was 371

shown to be dependent on NT5C3 expression (Fig. 4B). NT5C3 belongs to the 372

5’-nucleotidases family which mainly catalyze the dephosphorylation of pyrimidine 373

nucleoside monophosphates (2), which did not show any obvious effect on HBV 374

replication (data not shown). Interestingly, in a recent study, miR-122 level was found 375

to be negatively correlated with the expression of IRGs in patients with hepatitis C 376

(31). The collective results substantiate that IFN-α-induced ISG may act as a sponge 377

to inhibit miR-122. 378

We further explored the significance of IFN-α-induced miR-122 inhibition on 379

HBV infection. We found that miR-122 acts as an HBV suppressor miRNA, 380

indicating that IFN-α-induced miR-122 inhibition may attenuate the anti-HBV 381

activity of IFN-α. IFN-α is capable of inducing hundreds of genes (ISGs), and their 382

anti-viral effects have been extensively studied (21, 30, 16, 32, 3). However, the exact 383

role of each ISG still remains unclear and even controversial (39, 14). Contrary to the 384

currently accepted paradigm, we here present evidence to suggest that at least one ISG 385

NT5C3, which bears a matched miR-122 target sequence in the 3’-UTR of its mRNA, 386

sequesters and inhibits the HBV suppressor miR-122 by acting as a sponge, thus 387

induced NT5C3 by IFN-α may indirectly enhance HBV replication. Indeed, increased 388

HBV replication was observed under overexpression of the 3’-UTR of NT5C3 (Fig. 389

14

6A). Conceivably, other ISGs or IFN-α-regulated pathways may underlie the marked 390

suppression of HBV by IFN-α. 391

IFN-α therapy generates sustained virological response in a minority of CHB 392

patients (23, 35, 5). A recent study has shown that suppression of HBV replication by 393

IFN-α/β is dependent on viral load (38). These intriguing observations imply a 394

reciprocal effect of IFN-α on HBV replication. Conceivably, the regulation of 395

miR-122 by IFN-α may be one of the contributing reasons for the IFN-α-generated 396

different results. Hence, we believe that our current study not only raises potential 397

interest in increasing miR-122 to enhance the anti-HBV efficacy of IFN-α, but also 398

adds miR-122 as a possible marker for predicating the outcome of IFN therapy, as a 399

significant difference in miR-122 expression is observed among healthy individuals, 400

as well as in patients with CHB, CHC or HCC (6, 31, 42). 401

Finally, HCV and HBV that cause chronic infection, are notorious for their 402

capability to evade host defense and use cellular pathways for the establishment of 403

persistent infection. For HCV, this is the case as miR-122 has been shown to act in an 404

unusual manner to stimulate the replication and expression of HCV (17, 19). However, 405

this is not the case for HBV as we have shown that the most abundant liver-specific 406

miRNA miR-122 actually inhibits hepatotropic HBV replication. From an 407

evolutionary prospective, this intriguing phenomenon deserves further investigation. 408

In conclusion, our work presented herein has identified at least one ISG that 409

sequesters endogenous miR-122 by its mRNA 3’-UTR harboring the 410

miR-122-binding site, which may contribute to the IFN-α-mediated inhibition of 411

miR-122. Moreover, we provide evidence suggesting that miR-122 inhibition by 412

IFN-α attenuates the anti-virus effect of IFN-α as miR-122 leads to pronounced 413

inhibition of HBV replication. Given the broad function of miR-122 in the liver and 414

its anti-HBV activity, our work provides valuable insight for better understanding of 415

IFN-α-elicited anti-viral responses against HBV, and raises further potential interest 416

in enhancing the anti-HBV efficacy of IFN-α by increasing miR-122 expression. 417

418

Acknowledgements 419

15

We thank Fulian Liao and Yanlu Zan for technical help and advices in cell culture 420

and Dr. Wenlin Huang for kindly providing pRL-TK plasmid. 421

This work was supported by grants from the National Natural Science Foundation 422

of China (30970146, 31230026, 91029724, 81021003, 81102018). 423

424

References 425

426

1. Abe A, Inoue K, Tanaka T, Kato J, Kajiyama N, Kawaguchi R, Tanaka S, 427

Yoshiba M, Kohara M. 1999. Quantitation of hepatitis B virus genomic DNA by 428

real-time detection PCR. J Clin Microbiol. 37(9):2899-903. 429

2. Aksoy P, Zhu MJ, Kalari KR, Moon I, Pelleymounter LL, Eckloff BW, 430

Wieben ED, Yee VC, Weinshilboum RM, Wang L. 2009. Cytosolic 431

5'-nucleotidase III (NT5C3): gene sequence variation and functional genomics. 432

Pharmacogenet Genomics. 19(8):567-76. 433

3. Bonvin M, Achermann F, Greeve I, Stroka D, Keogh A, Inderbitzin D, 434

Candinas D, Sommer P, Wain-Hobson S, Vartanian JP, Greeve J. 2006. 435

Interferon-inducible expression of APOBEC3 editing enzymes in human 436

hepatocytes and inhibition of hepatitis B virus replication. Hepatology. 437

43(6):1364-74 438

4. Boutz DR, Collins PJ, Suresh U, Lu M, Ramírez CM, Fernández-Hernando 439

C, Huang Y, Abreu Rde S, Le SY, Shapiro BA, Liu AM, Luk JM, Aldred SF, 440

Trinklein ND, Marcotte EM, Penalva LO. 2011. Two-tiered Approach 441

Identifies a Network of Cancer and Liver Disease-related Genes Regulated by 442

miR-122. J Biol Chem. 286:18066-18078. 443

5. Buster EH, Hansen BE, Lau GK, Piratvisuth T, Zeuzem S, Steyerberg EW, 444

Janssen HL. 2009. Factors that predict response of patients with hepatitis B e 445

antigen-positive chronic hepatitis B to peginterferon-alfa. Gastroenterology. 446

137(6):2002-9. 447

6. Ebert MS, Neilson JR, Sharp PA. 2007. MicroRNA sponges: competitive 448

inhibitors of small RNAs in mammalian cells. Nat Methods. 4(9):721-6. 449

7. Ebert MS, Sharp PA. 2010. MicroRNA sponges: progress and possibilities. 450

RNA. 16(11):2043-50. 451

8. Filipowicz W, Grosshans H. 2001.The liver-specific microRNA miR-122: 452

biology and therapeutic potential. Prog Drug Res. 67:221-38. 453

9. Gatfield, D., G. Le Martelot, C. E. Vejnar, D. Gerlach, O. Schaad, F. 454

Fleury-Olela, A. L. Ruskeepaa, M. Oresic, C. C. Esau, E. M. Zdobnov, and 455

U. Schibler. 2009. Integration of microRNA miR-122 in hepatic circadian gene 456

expression. Genes Dev. 23:1313-26. 457 10. Girard M, Jacquemin E, Munnich A, Lyonnet S, Henrion-Caude. 2008. A 458

miR-122, a paradigm for the role of microRNAs in the liver. J Hepatol. 48: 459

648-656. 460

16

11. Gramantieri L, Ferracin M, Fornari F, Veronese A, Sabbioni S, Liu CG, 461

Calin GA, Giovannini C, Ferrazzi E, Grazi GL, Croce CM, Bolondi L, 462

Negrini M. 2007. Cyclin G1 is a target of miR-122a, a microRNA frequently 463

down-regulated in human hepatocellular carcinoma. Cancer Res. 67: 6092-6099. 464

12. He XS, Nanda S, Ji X, Calderon-Rodriguez GM, Greenberg HB, Liang 465

TJ. 2010. Differential transcriptional responses to interferon-alpha and 466

interferon-gamma in primary human hepatocytes. J Interferon Cytokine Res. 467

30(5):311-20. 468

13. Hsieh JL, Wu CL, Lee CH, Shiau AL. 2003. Hepatitis B Virus X Protein 469

Sensitizes Hepatocellular Carcinoma Cells to Cytolysis Induced by E1B-deleted 470

Adenovirus through the Disruption of p53 Function. Clin Cancer Res. 471

9(1):338-45. 472

14. Jost S, Turelli P, Mangeat B, Protzer U, Trono D. 2007. Induction of antiviral 473

cytidine deaminases does not explain the inhibition of hepatitis B virus 474

replication by interferons. J Virol. 81(19):10588-96. 475

15. Kelly EJ, Hadac EM, Cullen BR, Russell SJ. 2010. MicroRNA antagonism of 476

the picornaviral life cycle: alternative mechanisms of interference. PLoS Pathog. 477

6(3):e1000820. 478

16. Kim JH, Luo JK, Zhang DE. 2008. The level of hepatitis B virus replication is 479

not affected by protein ISG15 modification but is reduced by inhibition of UBP43 480

(USP18) expression. J Immunol. 181(9):6467-72. 481

17. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M, Munk 482

ME, Kauppinen S, Ørum H. 2010. Therapeutic silencing of microRNA-122 in 483

primates with chronic hepatitis C virus infection. Science. 327:198-201. 484

18. Li, C., S. Cao, Z. Liu, X. Ye, L. Chen, and S. Meng. 2010. RNAi-mediated 485

downregulation of uPAR synergizes with targeting of HER2 through the ERK 486

pathway in breast cancer cells. Int J Cancer. Oct 1. 127(7):1507-16. 487

19. Li YP, Gottwein JM, Scheel TK, Jensen TB, Bukh J. 2011. MicroRNA-122 488

antagonism against hepatitis C virus genotypes 1-6 and reduced efficacy by host 489

RNA insertion or mutations in the HCV 5' UTR. Proc Natl Acad Sci U S A . 490

108:4991-4996. 491

20. Lütgehetmann M, Bornscheuer T, Volz T, Allweiss L, Bockmann JH, Pollok 492

JM, Lohse AW, Petersen J, Dandri M. 2011. Hepatitis B virus limits response 493

of human hepatocytes to interferon-α in chimeric mice. Gastroenterology. Jun. 494

140(7):2074-83, 2083.e1-2. 495

21. Mao R, Zhang J, Jiang D, Cai D, Levy JM, Cuconati A, Block TM, Guo JT, 496

Guo H. 2011. Indoleamine 2,3-dioxygenase mediates the antiviral effect of 497

gamma interferon against hepatitis B virus in human hepatocyte-derived cells. J 498

Virol. 85(2):1048-57. 499

22. Moore KJ, Rayner KJ, Suárez Y, Fernández-Hernando C. 2011.The role of 500

microRNAs in cholesterol efflux and hepatic lipid metabolism.Annu Rev Nutr. 501

31:49-63. 502

23. Moucari R, Mackiewicz V, Lada O, Ripault MP, Castelnau C, 503

Martinot-Peignoux M, Dauvergne A, Asselah T, Boyer N, Bedossa P, Valla D, 504

17

Vidaud M, Nicolas-Chanoine MH, Marcellin P. 2009. Early serum HBsAg 505

drop: a strong predictor of sustained virological response to pegylated interferon 506

alfa-2a in HBeAg-negative patients. Hepatology. 49(4):1151-7. 507

24. Neuveut C, Wei Y, Buendia MA. 2010. Mechanisms of HBV-related 508

hepatocarcinogenesis. J Hepatol. 52: 594-604. 509

25. Norman KL, Sarnow P. 2010. Modulation of hepatitis C virus RNA abundance 510

and the isoprenoid biosynthesis pathway by microRNA miR-122 involves distinct 511

mechanisms. J Virol. 84:666-670. 512

26. Ono SK, Kato N, Shiratori Y, Kato J, Goto T, Schinazi RF, Carrilho FJ, 513

Omata M. 2001. The polymerase L528M mutation cooperates with nucleotide 514

binding-site mutations, increasing hepatitis B virus replication and drug resistance. 515

J Clin Invest. 107:449-455. 516

27. Pedersen IM, Cheng G, Wieland S, Volinia S, Croce CM, Chisari FV, David 517

M. 2007. Interferon modulation of cellular microRNAs as an antiviral mechanism. 518

Nature. 449(7164):919-22. 519

28. Qiu L, Fan H, Jin W, Zhao B, Wang Y, Ju Y, Chen L, Chen Y, Duan Z, Meng S. 520

2010. MiR-122-induced down-regulation of HO-1 negatively affects 521

miR-122-mediated suppression of HBV. Biochem Biophys Res Commun. 522

398:771-777. 523

29. Rudin, D., S. M. Shah, A. Kiss, R. V. Wetz, and V. M. Sottile. 2007. Interferon 524

and lamivudine vs. interferon for hepatitis B e antigen-positive hepatitis B 525

treatment: meta-analysis of randomized controlled trials. Liver Int. 27:1185-93. 526

30. Sadler, A. J., and B. R. Williams. 2008. Interferon-inducible antiviral effectors. 527

Nat Rev Immunol. 8:559-68. 528

31. Sarasin-Filipowicz, M., J. Krol, I. Markiewicz, M. H. Heim, and W. 529

Filipowicz. 2009. Decreased levels of microRNA miR-122 in individuals with 530

hepatitis C responding poorly to interferon therapy. Nat Med. 15:31-3. 531

32. Sarkis PT, Ying S, Xu R, Yu XF. 2006. STAT1-independent cell type-specific 532

regulation of antiviral APOBEC3G by IFN-alpha. J Immunol. 177(7):4530-40. 533

33. Shimizu S, Seki N, Sugimoto T, Horiguchi S, Tanzawa H, Hanazawa T, 534

Okamoto Y. 2007. Identification of molecular targets in head and neck squamous 535

cell carcinomas based on genome-wide gene expression profiling. Oncol Rep. 536

18(6):1489-97. 537

34. Shlomai, A., N. Paran, and Y. Shaul. 2006. PGC-1alpha controls hepatitis B 538

virus through nutritional signals. Proc Natl Acad Sci U S A. 103:16003-8. 539

35. Sonneveld MJ, Rijckborst V, Boucher CA, Hansen BE, Janssen HL. 2010. 540

Prediction of sustained response to peginterferon alfa-2b for hepatitis B e 541

antigen-positive chronic hepatitis B using on-treatment hepatitis B surface 542

antigen decline. Hepatology. 52(4):1251-7. 543

36. Tang, H., and A. McLachlan. 2001. Transcriptional regulation of hepatitis B 544

virus by nuclear hormone receptors is a critical determinant of viral tropism. Proc 545

Natl Acad Sci U S A. 98:1841-6. 546

18

37. Ter Borg, M. J., B. E. Hansen, G. Bigot, B. L. Haagmans, and H. L. Janssen. 547

2008. ALT and viral load decline during PEG-IFN alpha-2b treatment for 548

HBeAg-positive chronic hepatitis B. J Clin Virol. 42:160-4. 549

38. Tian Y, Chen WL, Ou JH. 2011. Effects of interferon-α/β on HBV replication 550

determined by viral load. PLoS Pathog. 7(7):e1002159. 551

39. Turelli P, Liagre-Quazzola A, Mangeat B, Verp S, Jost S, Trono D. 2008. 552

APOBEC3-independent interferon-induced viral clearance in hepatitis B virus 553

transgenic mice. J Virol. 82(13):6585-90. 554

40. Vivekanandan P, Thomas D, Torbenson M. 2009. Methylation regulates 555

hepatitis B viral protein expression. J Infect Dis. 199(9):1286-91. 556

41. Wang LY, Li YG, Chen K, Li K, Qu JL, Qin DD, Tang H. 2012. Stable 557

expression and integrated hepatitis B virus genome in a human hepatoma cell line. 558

Genet Mol Res. 11(2):1442-8. 559

42. Wang S, Qiu L, Yan X, Jin W, Wang Y, Chen L, Wu E, Ye X, Gao GF, Wang 560

F, Chen Y, Duan Z, Meng S. 2012. Loss of MiR-122 expression in patients with 561

hepatitis B enhances hepatitis B virus replication through cyclin G1 modulated 562

P53 activity. Hepatology. 55(3):730-41. 563

43. Wu M, Xu Y, Lin S, Zhang X, Xiang L, Yuan Z. 2007. Hepatitis B virus 564

polymerase inhibits the interferon-inducible MyD88 promoter by blocking 565

nuclear translocation of Stat1. J Gen Virol. 88(Pt 12):3260-9. 566

44. Xu C, Guo H, Pan XB, Mao R, Yu W, Xu X, Wei L, Chang J, Block TM, Guo 567

JT. 2010. Interferons accelerate decay of replication-competent nucleocapsids of 568

hepatitis B virus. J Virol. 84(18):9332-40. 569

45. Xu H, He JH, Xiao ZD, Zhang QQ, Chen YQ, Zhou H, Qu LH. 2010. 570

Liver-enriched transcription factors regulate microRNA-122 that targets CUTL1 571

during liver development. Hepatology. 52(4):1431-42. 572

46. Yang F, Zhang L, Wang F, Wang Y, Huo XS, Yin YX, Wang YQ, Zhang L, 573

Sun SH. 2011. Modulation of the unfolded protein response is the core of 574

microRNA-122-involved sensitivity to chemotherapy in hepatocellular carcinoma. 575

Neoplasia. 13(7):590-600. 576

47. Yang, J., and L. S. Zhao. 2009. Clinical significance of 4 patients with chronic 577

hepatitis B achieving HBsAg clearance by treated with pegylated interferon 578

alpha-2a for less than 1 year: a short report. Virol J. 6:97. 579

48. Y. Shan, J. Zheng, R.W. Lambrecht, H.L. 2007. Bonkovsky, Reciprocal effects 580

of micro-RNA-122 on expression of heme oxygenase-1 and hepatitis C virus 581

genes in human hepatocytes, Gastroenterology. 133(4):1166-74. 582

49. Zeng C, Wang R, Li D, Lin XJ, Wei QK, Yuan Y, Wang Q, Chen W, Zhuang 583

SM. 2010 . A novel GSK-3 beta-C/EBP alpha-miR-122-insulin-like growth factor 584

1 receptor regulatory circuitry in human hepatocellular carcinoma. Hepatology. 585

52(5):1702-12. 586

50. Zhang, Q., Y. Wang, L. Wei, D. Jiang, J. H. Wang, H. Y. Rao, L. Zhu, H. 587

Chen, R. Fei, and X. Cong. 2008. Role of ISGF3 in modulating the anti-hepatitis 588

B virus activity of interferon-alpha in vitro. J Gastroenterol Hepatol . 23:1747-61. 589

19

51. Zhang, S. Y., S. Boisson-Dupuis, A. Chapgier, K. Yang, J. Bustamante, A. 590

Puel, C. Picard, L. Abel, E. Jouanguy, and J. L. Casanova. 2008. Inborn errors 591

of interferon (IFN)-mediated immunity in humans: insights into the respective 592

roles of IFN-alpha/beta, IFN-gamma, and IFN-lambda in host defense. Immunol 593

Rev. 226:29-40. 594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

20

634

635

636

Figure legends: 637

Fig. 1. MiR-122 expression and HBV replication were suppressed by interferon-α 638

treatment. (A) Dose-response analysis of miR-122 induced by IFN-α. Huh-7 cells 639

were treated with the indicated concentration of IFN-α for 4 hr, and miR-122 640

expression was quantified with real-time PCR. The results were normalized to a U6 641

endogenous control RNU6B. (B, C and D) Time course of miR-122 expression in 642

response to IFN-α. Huh-7 cells were treated with 1000 U/ml of IFN-α once, and 643

miR-122 expression was quantified by real-time PCR at the indicated time (B). Huh-7 644

cells were treated with 1000 U/ml of IFN-α for every 12 hr, and miR-122 levels were 645

quantified by real-time PCR (C) and Northern blot (D) analysis at the indicated time. 646

(E-H) HBV expression and replication assays in the HBV replication plasmid pHBV 647

transfected cells under IFN-α treatment. Huh-7 cells were transfected with pHBV, and 648

4 hr later the cells were treated with 1000 U/ml of IFN-α for every 12 hr. The levels 649

of secreted HBsAg and HBeAg were quantified with ELISA and expressed as 650

O.D.450 nm (E). HBV DNA copies in the supernatant were quantified with real-time 651

PCR (F). HBV-DNA levels in cells were detected by Southern blot assay at 48 hr(G). 652

rcDNA, relaxed circular DNA; dsDNA, double-stranded DNA; ssDNA, 653

single-stranded DNA. HBV pgRNA and total RNA levels were measured by real-time 654

PCR (H). GAPDH was used as an internal control. The IFN-α-untreated cells served 655

as control. Error bars, means ± S.D. of three independent experiments. * P<0.05, ** 656

P<0.01 compared with control. 657

658

Fig. 2. Effect of interferon-α on miR-122 transcription. (A) Time course of miR-122 659

promoter activity in response to IFN-α treatment. Huh-7 cells were co-transfected 660

with pGL-122 with a luciferase reporter under miR-122 promoter and pRL-TK, and at 661

18 hr after transfection cells were treated with 1000 U/ml of IFN-α for the indicated 662

time. pGL-122 and Renilla luciferase activities were measured using dual luciferase 663

assay. (B) MiR-122 primary transcript (pri-miR-122) was quantified by real-time PCR 664

21

in Huh-7 cells treated with 1000 U/ml of IFN-α for the indicated time. The 665

IFN-α-untreated cells served as control. Error bars, means ± S.D. from three 666

independent experiments. * P<0.05, ** P<0.01 compared with control. 667

668

Fig. 3. Inhibition of miR-122 by its target mRNA of NT5C3 induced by interferon-α. 669

(A) Confirmation of NT5C3 as the miR-122 target by luciferase reporter assay. 670

Perfect matches are indicated by a line showing predicted binding sequence of 671

miR-122 within the 3’-UTR of NT5C3 mRNA. Mutations are made to the seed region 672

of the miR-122-binding site (NT5C3-mut) for the reporter gene assay. Huh-7 cells 673

were co-transfected with a miR-122 mimic or miR-152 or miR-642 or a randomized 674

oligonucleotide as mock, and a firefly luciferase reporter plasmid carrying either the 675

wild-type (NT5C3-wt) or mutant (NT5C3-mut) 3’-UTR of NT5C3. At 48 hr 676

post-transfection, firefly luciferase and Renilla luciferase activities were measured, 677

and the luciferase activity was normalized to Renilla luciferase. (B and C) The 678

3’-UTR of NT5C3 mRNA sequesters endogenous miR-122. Huh-7 cells were 679

transfected with the reporter plasmid with wild-type (NT5C3-wt) or mutant 680

(NT5C3-mut) 3’-UTR of NT5C3 or the empty plasmid pGL3 as control, and miR-122 681

levels were quantified by real-time PCR 24 and 48 hr after transfection,firefly 682

luciferase and Renilla luciferase activities were measured at 24 and 48hr , and the 683

luciferase activity was normalized to Renilla luciferase (B). The miR-122 levels at 24 684

hr were detected by Northern blot analysis (C). (D) HepG2 cells were transfected with 685

a miR-122 mimic or a randomized oligonucleotide as mock, and Huh-7 cells were 686

transfected with a miR-122 inhibitor or randomized oligonucleotide as mock. The 687

cylinG1 protein levels were measured by immunoblotting 48 hr post-transfection. 688

Actin was used as a loading control. (E) Huh-7 cells were transfected with the reporter 689

plasmid with wild-type (NT5C3-wt) or mutant (NT5C3-mut) 3’-UTR of NT5C3, and 690

the cyclinG1 protein levels were measured by immunoblotting 48 hr post-transfection. 691

Actin was used as a loading control. (F) Time course of NT5C3 mRNA expression in 692

response to IFN-α treatment. Huh-7 cells were treated with 1000 U/ml of IFN-α for 693

22

the indicated time, and NT5C3 mRNA levels were quantified by real-time PCR. Error 694

bars, means ± S.D. from three independent experiments. * P<0.05, ** P<0.01 695

compared with control or mock. 696

697

Fig. 4. Inhibition of miR-122 by interferon-α is dependent on NT5C3 mRNA. (A) 698

Huh-7 cells were transfected with the NT5C3-specific siRNA (siNT5C3) or a 699

scrambled siRNA as mock. The miR-122 levels were determined 24 hr 700

post-transfection by real-time PCR and Northern blot analysis (A). (B) Huh-7 cells 701

were transfected with siNT5C3 or a scrambled siRNA as mock, and at 20 hr after 702

transfection 1000 U/ml of IFN-α was added. NT5C3 mRNA levels and miR-122 703

levels were determined at 4 hr after IFN-α treatment by real-time PCR. Error bars, 704

means ± S.D. from three independent experiments. * P<0.05, ** P<0.01 compared 705

with mock. 706

707

Fig.5. MiR-122 inhibits HBV expression and replication. (A and B) Huh-7 cells were 708

co-transfected with pHBV and 25 nM of miR-122 inhibitor or 25 nM miR-152 or 709

miR-642 or a randomized oligonucleotide as mock. At 48 hr after transfection, the 710

secretion of HBsAg and HBeAg was measured by ELISA (A), and HBV DNA copies 711

in the supernatant were quantified by real-time PCR (B). (C-F) HepG2 cells were 712

co-transfected with pHBV and 50 nM of chemically synthesized miR-122 mimic or 713

50 nM miR-152 or miR-642 or a randomized oligonucleotide as mock. At 48 hr after 714

transfection, the secretion of HBsAg and HBeAg was measured by ELISA (C). HBV 715

total mRNA levels (D) and DNA copies (E) in the supernatant were quantified by 716

real-time PCR. (F) HBV-DNA levels in cells were detected by Southern blot assay. 717

Cells without transfection served as negative control for Southern blot. The cylinG1 718

protein levels were measured by immunoblotting. Actin was used as a loading control. 719

Student’s t-test was used to determine p-values. * P<0.05, ** P<0.01 compared with 720

mock. 721

722

Fig. 6. Abrogation of IFN-α-induced down-regulation of miR-122 results in enhanced 723

23

suppression of HBV by IFN-α. (A) Huh-7 cells were co-transfected with pHBV and a 724

luciferase reporter plasmid carrying either the wild-type (NT5C3-wt) or mutant 725

(NT5C3-mut) 3’-UTR of NT5C3. HBV DNA copies in the supernatant were 726

quantified by real-time PCR 48 hr after transfection. (B) Huh-7 cells were 727

co-transfected with pHBV and 25 nM of miR-122 mimic. At 6 hr after transfection 728

500 U/ml of IFN-α was added to the cell culture. HBV DNA copies in the supernatant 729

were measured by real-time PCR 48 hr post-transfection. (C) Huh-7 cells were 730

co-transfected with pHBV and 50 nM of miR-122 inhibitor. At 6 hr after transfection 731

1000 U/ml of IFN-α was added to the cell culture. HBV DNA copies in the 732

supernatant were measured by real-time PCR 48 hr post-transfection. 733

734

Fig.7. The mutant p53 in Huh-7 cells could regulate HBV transcription and 735

replication. (A and B) Huh-7 cells were transfected with 100 nM specific siRNA or a 736

scrambled siRNA as mock. The p53 mRNA and protein levels were determined 48 hr 737

post-transfection by real-time PCR (A) and Western blot analysis (B), respectively. (C) 738

Huh-7 cells were co-transfected with 1μg pHBV-luc and 50 nM p53 siRNA or a 739

scrambled siRNA as mock. At 48 hr post-transfection, firefly luciferase and Renilla 740

luciferase activities were measured, and the luciferase activity was normalized to 741

Renilla luciferase. (D and E) Huh-7 cells were co-transfected with pHBV and p53 742

siRNA or a scrambled siRNA as mock. At 48 hr after transfection, the levels of 743

HBsAg and HBeAg in the supernatant were measured by ELISA (D), and HBV DNA 744

copies in the supernatant were quantified by real-time PCR (E). Error bars, means ± 745

S.D. from three independent experiments. * P<0.05, ** P<0.01 compared with mock. 746