JVI Accepts, published online ahead of print on 10 December 2014 J. Virol. doi:10.1128/JVI.02760-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Hepatitis B virus polymerase disrupts K63-linked ubiquitination of STING to block

2

innate cytosolic DNA-sensing pathways

3 4

Yinghui Liu1*, Jianhua Li2*, Jieliang Chen2,Yaming Li2, Weixia Wang2, Xiaoting

5

Du2, Wuhui Song2, Wen Zhang2, Li Lin2, Zhenghong Yuan1,2#

6 7 8

Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China1; Key

9

Laboratory of Medical Molecular Virology, Shanghai Medical College, Fudan

10

University, Shanghai, PR China2.

11 12

Running Head: HBV polymerase blocks STING-mediated IFN induction

13

# Address correspondence to Zhenghong Yuan, [email protected].

14

*Y. L. and J. L. contributed equally to this work.

15 16

The word count for the abstract: 388 (238 in Abstact, 150 in Importance)

17

The word count for the text: 6337

18 19 20 21 22 23 24 25 26 27 28 29 30 1 / 32

31

Abstract

32

The cellular innate immune system recognizing pathogen infection is essential for

33

host defense against viruses. In parallel, viruses have developed a variety of strategies

34

to evade the innate immunity. The hepatitis B virus (HBV), a DNA virus that causes

35

chronic hepatitis, has been shown to inhibit the RNA helicase RIG-I-mediated

36

interferon (IFN) induction. However, it is still unknown whether HBV could affect

37

the host DNA-sensing pathways. Here we report that in transient HBV-transfected

38

Huh7 cells, HBV stably producing cell line HepAD38, and HBV-infected HepaRG

39

cells and primary human hepatocytes, HBV markedly interfered with IFN-β induction

40

and antiviral immunity mediated by STING, which has been identified as a central

41

factor in foreign DNA recognition and antiviral innate immunity. Screening analysis

42

demonstrated that the viral polymerase (Pol), but not other HBV-encoded proteins,

43

was able to inhibit STING-stimulated interferon regulatory factor 3 (IRF3) activation

44

and IFN-β induction. Moreover, the reverse transcriptase (RT) and the ribonuclease H

45

(RH) domains of Pol were identified to be responsible for the inhibitory effects.

46

Furthermore, Pol was shown to physically associate with STING and dramatically

47

decrease the K63-linked polyubiquitination of STING via its RT domain without

48

altering the expression level of STING. Taken together, these observations suggest

49

that besides its inherent catalytic function, Pol has a role in suppression of IFN-β

50

production by direct interaction with STING and subsequent disruption of its

51

K63-linked ubiquitination, providing a new mechanism for HBV to counteract the

52

innate DNA-sensing pathways.

53

Importance

54

Although whether and how HBV infection induces the innate immune responses is

55

still controversial, it becomes increasingly clear that HBV has developed strategies to

56

counteract the pattern recognition receptors-mediated signaling pathways. Previous

57

studies have shown that type I IFN induction activated by the host RNA sensors could

58

be inhibited by HBV. However, it remains unknown whether HBV as a DNA virus

59

utilizes evasion mechanisms against the foreign DNA-elicited antiviral signalings. In

60

recent years, the cytosolic DNA sensor and key adaptor STING has been 2 / 32

61

demonstrated to be essential in multiple foreign DNA-elicited innate immune

62

signalings. Here, for the first time, we reported STING as a new target of HBV to

63

antagonize the IFN induction and identified the viral polymerase responsible for the

64

inhibitory effect, thus providing an additional molecular mechanism by which HBV

65

evades the innate immunity and implying HBV polymerase is a multifunctional

66

immunomodulatory protein besides its inherent catalytic function.

67 68

Introduction

69

Hepatitis B virus(HBV) is one of the most important pathogens causing liver

70

diseases. Worldwide, approximately 350-400 million individuals are chronically

71

infected, many of whom are at increased risk of developing cirrhosis and

72

hepatocellular carcinoma (HCC)(1, 2). Although the underlying mechanisms leading

73

to chronic HBV infection remain to be clearly defined, the outcome of HBV infection

74

is thought to be the results of complex interactions between replicating HBV and the

75

host immune system (3).

76

The innate immunity constitutes the first line of defense against invading pathogens,

77

which recognizes the pathogen-associated molecular patterns (PAMPs) through

78

germline-encoded pattern recognition receptors (PRRs). Viral infection usually

79

activates some PRR(s), leading to type I interferons (IFNs, including IFN-α and

80

IFN-β) and inflammatory activities (4, 5). However, viruses including HBV have

81

developed a variety of strategies to counteract the host immune responses for the

82

benefit of their survival. It is reported that HBV surface antigen (HBs), e antigen

83

(HBeAg) and HBV virions could inhibit Toll-like receptors (TLRs)-mediated

84

production of type I IFN and pro-inflammatory cytokines in murine liver cells (6). In

85

addition, HBV x protein (HBx) was reported to negatively regulate retinoic

86

acid-inducible gene I (RIG-I)-mediated antiviral responses (7-9), while the viral

87

polymerase (Pol) was shown to suppress type I IFN induction through impairing

88

RIG-I- and TLR3-stimulated signalings (10, 11), both of which are RNA-sensing

89

pathways. Considering that HBV is a DNA-containing virus with a genome size of

90

3.2 Kb and there are at least two types of viral DNAs distinct from the host DNA 3 / 32

91

including relaxed circular DNA (rcDNA) and covalently closed circular DNA

92

(cccDNA) during its life cycle, we thus speculate that HBV may also have strategies

93

to interfere with the host DNA-sensing pathways.

94

Significant progress has been made in recent years in understanding how the innate

95

immune system detects non-self DNA molecules or DNA-containing pathogens.

96

Several proteins, including DNA-dependent activator of IFN-regulatory factors

97

(DAI)(12, 13), absent in melanoma 2 (AIM2) (14-16), the member of the PYHIN

98

protein family IFI16 (17), the member of the DEXDc family of helicases DDX41 (18),

99

the cGAMP synthase (cGAS) (19, 20) have been identified as DNA sensors.

100

Interestingly, the downstream signalings activated by most of these DNA sensors

101

converge on an essential signal transducer, the stimulator of interferon genes (STING,

102

also known as MITA, ERIS, TMEM173 and MPYS)(21-24). Meanwhile, STING is

103

reported as a direct innate immune sensor of cyclic diguanylate monophosphate

104

(c-di-GMP), a bacterial second messenger(25). Collectively, STING, functioning at

105

the signaling “traffic junction”, plays a critical role in the regulation of the immune

106

response to microbial nucleic acids, particularly the cytosolic DNA and cyclic

107

dinucleotides (CDNs). However, little is known about whether and how HBV disturbs

108

STING signaling in human hepatocytes.

109

Therefore, the aim of this study was to investigate the possible impact of HBV

110

replication on the STING-mediated type I IFN induction pathway. The results

111

revealed a novel mechanism employed by HBV to escape the innate immunity and

112

provided evidence for a new role of the viral polymerase in the inhibition of IFN-β

113

production through disrupting the K63-linked ubiquitination of STING.

114

Materials and Methods

115

Plasmids and viruses

116

The following expression plasmids were generously provided: pIFN-β-Luc (Rongtuan

117

Lin, McGill University, Canada); pEF-STING, and p-55C1B-Luc (Takashi Fujita,

118

Kyoto University, Japan); pFlag-MITA, pcDNA3.1-HA-MITA, pFlag-TRIM32 and

119

pFlag-TRIM56

120

pcDNA3.1-CMV-HBV1.1 (Youhua Xie, Fudan University, China) which contains the

(Hongbing

Shu, 4 / 32

Wuhan

University,

China);

121

wild-type HBV 1.1-mer overlength genomic sequence. pHBV1.3, pFlag-Pol

122

(pcDNA3.1-Flag-Pol and pQCXIP-Flag-Pol), the series of truncated mutants of Pol,

123

pcDNA3.1-Flag-Core,

pcDNA3.1-Flag-HBx,

pcDNA3.1-Flag-Precore,

124

pcDNA3.1-Flag-HBsAg,

pFlag-TBK1,pHA-RIG-I,

pCMV-Myc-Pol

125

pcDNA3-HA-Ub were described previously (10, 26-29). pHBV1.3-ΔPol and

126

pCMV-HBV-ΔPol, defective in viral polymerase expression, were derived from

127

pHBV1.3 and pcDNA3.1-CMV-HBV1.1 respectively by introducing a frameshift

128

mutation into the Pol gene after codon 108. The mutation strategy was described

129

previously(30). pHBV1.3-YMHD and pFlag-Pol-YMHD, lacking the polymerase

130

catalytic active site, were derived from pHBV1.3 and pFlag-Pol, respectively by

131

introducing a substitution mutation into the Pol gene. The amino acid was exchanged

132

from

133

GGCTTTCAGTTATATGCATGATGTGGT (the mutated nucleotide was underlined).

134

The mutation strategy was described previously (31). The carboxy-terminally

135

Flag-tagged Pol pPol-Flag was constructed by inserting a DNA fragment encoding

136

three repeats of the flag epitope tag at the carboxy terminus of Pol on the pcDNA3.1

137

vector.

138

The mutants pHA-Ub-K48R and pHA-Ub-K63 were derived from pcDNA3-HA-Ub.

139

In mutant pHA-Ub-K48R, the lysine at position 48 was substituted with arginine by in

140

vitro mutagenesis. In pHA-Ub-K63, all lysines except that at position63 (including

141

the lysines at positions 6, 11, 27, 29, 33 and 48) were mutated to arginines. pRL-TK

142

was purchased from Promega. NDV-GFP (a gift from Dr Yan Yuan, University of

143

Pennsylvania,

144

specific-pathogen-free-maintained chicken eggs.

145

Antibodies and reagents

146

Anti-β-actin, anti-Flag M2, anti-GFP and anti-HA antibodies were purchased from

147

Sigma-Aldrich. Anti-c-Myc and anti-IRF3 (FL425) antibodies were obtained from

148

Santa Cruz Biotechnology. Anti-STING antibody was obtained from Proteintech.

149

Anti-Core, anti-HBs and anti-Ub antibodies were purchased from Dako, Shanghai

150

Long Island Biotech and Covance, respectively. Anti-TRIM32 antibody was a gift

YMDD

to

YMHD

PA,

in

USA)

the

polymerase

was

propagated

5 / 32

sequence.

and

The

and

primer

purified

is

from

151

from Dr. Hongbing Shu (Wuhan University, China). The proteasome inhibitor

152

MG-132 was purchased from Calbiochem. cGAMP and polydA:dT were purchased

153

from Biolog and Amersham Biosciences, respectively.

154

Cell culture and transfection

155

The cell lines HEK293, HEK293T, Vero(obtained from the Cell Bank of the Chinese

156

Academy of Science (Shanghai, China)) and Huh7 were cultured in Dulbecco’s

157

modified Eagle’s medium supplemented with 10% fetal bovine serum (FBS; Gibco),

158

penicillin (100 IU ml-1; Gibco) and streptomycin(100 μg ml-1; Gibco), in a 5% CO2

159

atmosphere at 37℃. The HepG2-derived HBV-producing stable cell line HepAD38

160

which was a kind gift from Yumei Wen, was maintained as described previously (32).

161

Additionally, HepAD38 cells were grown in presence or absence of 1 μg ml-1

162

doxycycline (Dox) to regulate HBV pregenomic RNA transcription. PH5CH8, a

163

simian virus 40 large T antigen-immortalized non-neoplastic human hepatic cell line

164

with intact capacity for type I IFN induction was maintained as described previously

165

(33). HepaRG cells (34) were purchased from BIOPREDIC INTERNATIONAL,

166

France. To obtain the differentiated HepaRG (dHepaRG), the cells were cultured for

167

two weeks in standard medium, then for two more weeks in medium supplemented

168

with 1.8% dimethylsulfoxide according to the manufacturers’ instructions. Primary

169

human hepatocytes (PHHs) were purchased from Shanghai RILD Inc. (Shanghai,

170

China). The cell culture was performed as described previously with slight

171

modification(35).The cell pellet containing PHHs was resuspended in the plating

172

medium of Williams E medium supplemented with 10% FBS 5 μg ml-1 transferrin, 5

173

ng ml-1 sodium selenite, 3μg ml-1 insulin, 2 mM L-glutamine, 100 U ml-1 penicillin,

174

100 μg ml-1 streptomycin. The cells were plated in the collagen I pre-coated chambers

175

(Lab-Tek) or the cell culture plates. 5 hr after plating, medium were changed to pri-

176

mary hepatocytes maintenance medium (PMM), that is, Williams E medium

177

supplemented with 5% FBS, 5 μg ml-1 transferrin, 10 ng ml-1 EGF, 3 μg ml-1 insulin, 2

178

mM L-glutamine, 18 μg ml-1 hydrocortisone, 40 ng ml-1 dexamethasone, 5 ng ml-1

179

sodium selenite, 2% DMSO, 100 U ml-1 penicillin,100 μg ml-1 streptomycin.

180

Transient transfection was performed with the indicated plasmids using Fugene HP 6 / 32

181

(Roche) or Lipofectamine 2000 (Invitrogen), according to the manufacturer’s

182

instructions. Digitonin permeabilization was utilized to deliver cGAMP into cultured

183

cells as described previously (36).

184

HBV infection of HepaRGs and PHHs

185

HBV infection of the hepatocytes was performed as described previously with slight

186

modification (37). The dHepaRG cells or PHHs were incubated overnight with

187

HBV-positive sera at approximate 100 genome equivalent copies of HBV per cell (1

188

vol of infectious pooled serum from 10 CHB patients diluted in 10 vol of culture

189

medium) containing 4% PEG 8000.After incubation, cells were rinsed three times.

190

The HBeAg secretion and the viral DNA in the medium were determined every two

191

or three days post-infection (data not shown). The cells successfully infected with

192

HBV were processed to further treatment and experiments at least nine days

193

post-infection.

194

The HBV-positive sera were collected from CHB patients in Shanghai Public Health

195

Clinical Center with informed consent and the approval of the institutional ethics

196

committee.

197

Dual-luciferase reporter assay

198

Cells (1×105) seeded in a 24-well plate were cultured overnight and then transfected

199

with the indicated stimulator plasmid (such as pSTING), reporter plasmid (pRL-TK,

200

pIFN-β-Luc or p-55C1B-Luc ) and the indicated plasmids using either Lipofectamine

201

2000 (Invitrogen) or Fugene HP (Roche) according to the manufacturer’s instructions.

202

At 36h post transfection, cells were lysed with passive lysis buffer and assayed using

203

a Dual-Luciferase Assay kit (Promega). Data were processed as described previously

204

(10). Data are expressed as the mean fold induction ± SD relative to control levels.

205

The results are representative of three independent experiments, each performed in

206

triplicate.

207

Real-time RT-PCR

208

Total cellular RNA was extracted with TRIzol reagent (Invitrogen), and then

209

subjected to reverse-transcription using

210

manufacturer’s instructions. The cDNA samples were subjected to real-time PCR 7 / 32

the

Kit

(TaKaRa)

following

the

211

using primers specific for human IFN-β or human interferon stimulated gene

212

56(ISG56) and human glyceraldehyde 3-phosphate dehydrogenase (GAPDH): IFN-β

213

forward,

214

5′-CTTCAGGTAATGCAGAATCC-3′;

215

5′-TAGCCAACATGTCCTCACAGAC-3′,and

216

5′-TCTTCTACCACTGGTTTCATGC-3′;

217

5′-GGTATCGTGGAAGGACTCATGA-3′,

218

5′-ATGCCAGTGGCTTCCCGTTCAGC-3′. For comparisons, transcription of

219

IFN-β and ISG56 was normalized to that of GAPDH. Data are expressed as the mean

220

fold induction ± SD relative to control levels. The results are representative of three

221

independent experiments, each performed in triplicate.

222

IFN-β ELISA

223

The level of IFN-β in the culture medium was measured using an ELISA kit for

224

human IFN-β (PBL Interferon Source) according to the manufacturer’s instructions.

225

Immunoprecipitation and Western blot analysis

226

For immunoprecipitation, HEK293T (1×107) or HEK293 (4×107) cells were

227

transfected with various combinations of plasmids using Lipofectamine 2000

228

(Invitrogen) for 48 h and then lysed in immunoprecipitation buffer(50 mM Tris-HCl

229

pH7.5, 150 mM NaCl, 0.5% (vol/vol) NP-40, 10% glycerol, 1mM EDTA)and

230

supplemented with a protease inhibitor cocktail (Roche). Cell debris was removed by

231

centrifugation at 12,000 g for 10 min at 4°C. The supernatants were collected,

232

precleared with protein A/G Plus agarose beads (Santa Cruz) for 30min , and

233

incubated with an anti-FLAG or anti-HA antibodies at 4°C. After 2 h, 25μl of 1:1

234

slurry of protein A/G Plus agarose beads was added and incubated for additional 2h.

235

The immunoprecipitates were washed five times with lysis buffer and boiled in 1%

236

(wt/vol) SDS sample buffer, followed by SDS-PAGE and Western blot analysis as

237

described previously (27).

238

Confocal microscopy

239

Immunofluorescence was performed as described (10). The fluorescence was

240

observed under a confocal fluorescence microscope (Leica TCS SP2).

5′-GATTCATCTAGCACTGGCTGG-3′,and

8 / 32

ISG56

reverse, forward reverse,

GAPDH and

forward, reverse,

241

Southern blot analysis

242

Intracellular HBV core particle-associated DNA in Huh7 cells was extracted at 48h

243

post-transfection and Southern blot analysis was performed as described previously

244

(29).

245

Native PAGE

246

For the detection of IRF3 dimerization, 3×105 HEK293 cells were seeded into a

247

12-well plate, cultured overnight and then transfected with the indicated amounts of

248

pEF-STING and pQCXIP-Flag-Pol (empty vector was used to balance the total

249

amount of DNA) using Lipofectamine 2000. After 24 h, cells were selected with

250

puromycin (1.5μg ml-1) for 36 h. The cells were harvested with 40μl ice-cold lysis

251

buffer (50 mM Tris/HCl, (pH 7.5), 150 mM NaCl and 0.5% NP-40 containing 1×

252

Roche protease inhibitors). After centrifugation at 13000 g for 10 min, supernatants

253

were quantified using a BCA assay (Thermo Scientific) and diluted with 5×native

254

PAGE sample buffer (312.5 mM Tris/HCl (pH 6.8), 75% glycerol and 0.25%

255

bromophenol blue), and then 60 μg of total protein was applied to a pre-runned 6%

256

native gel for separation. After electrophoresis, the proteins were transferred onto a

257

nitrocellulose membrane for Western blot.

258

For the detection of HBV core capsids, cells were harvested with the above lysis

259

buffer at 36h post-transfection and then centrifugated at 13000 g for 10 min. The

260

quantified supernatants were subjected to a native agarose gel, blotted onto a

261

nitrocellulose membrane, and detected for the formation of core capsids using the

262

anti-Core antibody.

263

In vivo ubiquitination assay

264

HEK293T cells in a 60-mm-diameter dish were co-transfected with the indicated

265

plasmids using Lipofectamine 2000 (Invitrogen). For the detection of STING

266

ubiquitination, at 36 h post transfection, cells were treated with the proteasome

267

inhibitor MG-132 at a final concentration of 20 μM for 6 h.The cells were washed

268

twice with PBS and lysed with ice-cold RIPA buffer (50 mM Tris-HCl (pH 7.5),

269

150mM NaCl, 1%NP-40, 0.5% Deoxycholic acid sodium, 0.1% SDS, supplemented

270

with a protease inhibitor cocktail (Roche) and 20μM MG132) for 5-10 min. Cell 9 / 32

271

debris was removed by centrifugation at 12,000 g for 10 min at 4°C. The supernatants

272

were divided into two aliquots. One aliquot (5%) was prepared for Western blot. The

273

second aliquot (95%) was sonicated five times to shear DNA. The soluble lysates

274

were then immunoprecipitated with anti-HA antibody followed by three washes with

275

RIPA buffer. HA-tagged proteins were resolved by SDS-PAGE and sequentially

276

blotted with the indicated antibodies.

277

Infection protection assay

278

Huh7 cells were seeded in a 24-well plate and transfected with the indicated plasmids.

279

After 36h, the cells were mock-treated or treated with 100nM cGAMP for 16h. The

280

supernatants were collected and filtered by 0.22μm filters (Millipore). Vero cells in

281

24-well plate were incubated with the filtered Huh7 supernatants overnight, and then

282

challenged with 80 haemagglutinin units (HAU) ml-1 of NDV-GFP for additional 24h,

283

followed by observation under fluorescent microscope and analysis by Western blot.

284

Statistical analysis

285

Data were analyzed using Student’s t-test and presented as the mean ± Standard

286

deviations. A value of P﹤0.05 was considered to be statistically significant.

287

Results

288

1. HBV inhibits STING-mediated IFN-β induction and antiviral immune

289

response.

290

To explore whether HBV could regulate STING-mediated type I IFN induction, we

291

first test the overexpressed STING-stimulated IFN-β promoter activation inHuh7 cells

292

transfected with two kinds of HBV replicative constructs, pHBV1.3 or pCMV-HBV

293

in which HBV replication is driven by either HBV native promoter or the more potent

294

cytomegalovirus (CMV) promoter, respectively. Meanwhile, we monitored HBV

295

replication level by detecting the formation of core capsids using native agarose gel.

296

As shown in Fig. 1A (left part), STING-mediated IFN-β promoter activation was

297

abrogated in a dose-dependent manner in both of the HBV replicative

298

plasmid-transfected cells, particularly in the cells transfected with pCMV-HBV in

299

which higher level of core capsids was detected (Fig.1A, right part). The interferon

300

regulatory factor 3 (IRF3), as a key transcriptional factor, is essential for 10 / 32

301

STING-stimulated signaling transduction and IFN-β induction (22, 24). To further

302

confirm the effect of HBV on STING-mediated IFN-β induction, the effect of HBV

303

on STING-mediated IRF3 activation was tested using the reporter plasmid

304

p-55C1B-Luc in which the luciferase expression was driven by a promoter containing

305

repeated IRF-binding sites. The results showed that HBV effectively inhibited the

306

STING-mediated IRF3 activation (Fig. 1B).

307

Cyclic GMP-AMP (cGAMP) has been identified as a specific upstream activator of

308

STING to induce type I IFN production (19, 20, 38, 39). Because STING expression

309

was barely detectable in Huh7 cells and HepaAD38 cells (Fig. 1C) as reported

310

previously (40), we co-transfected the cells with STING-encoding plasmids to

311

reconstitute the responsiveness of the cells to cGAMP when reporter plasmids and

312

plasmids containing HBV genome or control vectors were transfected into the cells.

313

Upon cGAMP stimulation, significant activation of IFN-β promoter was observed,

314

however, it was markedly impaired in the cells transfected with pHBV1.3 or

315

pCMV-HBV (Fig.1D).Moreover, the amount of cGAMP-triggered IFN- production

316

in Huh7 cells was also decreased in the presence of HBV replication in a

317

dose-dependent manner (Fig. 1E), indicating that the cGAMP-STING-mediated

318

signaling pathway was suppressed by HBV.

319

To further evaluate the inhibitory effect of HBV on the STING-mediated antiviral

320

response, we performed a virus infection protection assay, as described previously

321

(10). For this, the antiviral activities of supernatants harvested from IFN-α-or

322

cGAMP-treated Huh7 cells with or without HBV replication were assayed in Vero

323

cells challenged with the Newcastle disease viral particles tagged with green

324

fluorescent protein (NDV-GFP) by determination of the expression level of extent of

325

green fluorescence. The results showed that NDV-GFP expression was diminished in

326

Vero cells pre-treated with the supernatants from cGAMP- and IFN-α-treated Huh7

327

cells (Fig. 1F lanes/panels 4 and 8). In contrast, the supernatants from Huh7 cells

328

transfected with the pHBV1.3construct lost antiviral activities depending on the

329

transfected dose, as judged by dim green fluorescence (Fig. 1F lanes/panels 5,6 and 7),

330

implying that the innate antiviral response induced by cGAMP in Huh7 cells was 11 / 32

331

remarkably decreased in the presence of HBV.

332

To better understand the interference of HBV on STING-induced type I IFN

333

production in the HBV stably producing cell model, we tested the cGAMP-stimulated

334

IFN-β promoter activation in HepaAD38 cell line, which expresses HBV under the

335

control of an inducible tetracycline-off (Tet-off) promoter (32). The cells were

336

pre-transfected with the STING expressing plasmid before cGAMP stimulation for

337

the reasons mentioned above that the endogenous expression level of STING is rather

338

low in HepAD38 cells. As shown in Fig. 1G, both 100nM and 400nM of

339

cGAMP-stimulated IFN-β promoter activation was statistically lower, almost 35% in

340

the cells without doxycycline than those with doxycycline.

341

To further validate whether the suppression of HBV on STING signaling occuring

342

a natural infection of hepatocytes, differentiated HepaRG (dHepaRG) cells and PHHs

343

infected with HBV-positive sera were mock-treated or treated with cGAMP. In

344

dHepaRG cells, the immunofluorescece results showed that the IRF3 protein was

345

predominantly localized in the cytoplasm in the mock cells. Upon cGAMP

346

stimulation, IRF3 translocated into the nucleus in about 70% of hepatocyte-like cells,

347

but the nuclear translocation of IRF3 was significantly reduced in those HBV-infected

348

cells (using HBsAg as a positive marker for HBV infection)(Fig. 1H). In PHHs which

349

support a higher-efficient viral infection (about 30%-40%) (Fig. 1I, left part), the

350

cGAMP-elicited IFN-β gene production in HBV-infected cells was obviously lower

351

compared with that in uninfected cells (Fig. 1I, right part). The data strongly support

352

the results obtained in the overexpression system that the presence of HBV replication

353

dampened STING-activated signaling pathways.

354

Overall, these results indicated that HBV may have developed some strategies to

355

evade the STING-mediated host antiviral immune responses.

356

2. HBV polymerase (Pol) suppresses STING-induced IFN-β production.

357

To determine which viral proteins are responsible for the inhibition of

358

STING-mediated IFN-β induction by HBV, we performed a functional screening

359

assay by co-transfecting the constructs encoding HBV proteins, including polymerase

360

(Pol), pre-Core, Core, HBx, or HBs together with the STING-expressing plasmid and 12 / 32

361

the reporter plasmids into Huh7 cells. The results showed that only expression of

362

HBV Pol, but not other viral proteins, obviously suppressed the IFN-β promoter

363

activation induced by ectopic expression of STING in Huh7 cells (Fig.2A), and the

364

inhibitory effect was shown in a dose-dependent manner using both of the amino- and

365

carboxy-terminally Flag-tagged Pol constructs (Fig.2B). To further confirm the

366

suppression of Pol on the STING-mediated endogenous IFN-β production, HEK 293

367

cells were co-transfected with plasmids encoding HBV Pol and STING and the

368

cellular mRNA levels of IFN-β and interferon-stimulated gene 56 (ISG56) were

369

quantified by real-time qPCR. The results showed that the expression of Pol resulted

370

in decreased production of both IFN-β and ISG56 mRNAs (Fig.2C). We next

371

examined the effect of HBV polymerase on STING-mediated IRF3 activation in

372

HEK293 cells by detecting

373

hallmark of IRF3 activation and IFN induction (41). The results demonstrated that

374

STING-induced dimerization of IRF3 was inhibited by Pol in a dose-dependent

375

manner (Fig.2D).

the dimerization of IRF3 which is considered as a

376

We further confirmed the inhibitory effect of Pol on the STING-mediated IFN-β

377

induction using cGAMP and poly(dA:dT) as STING activators. First, we used

378

cGAMP as an IFN-β stimulus. Consistently, Pol also inhibited STING-mediated

379

IFN-β promoter activation upon cGAMP stimulation in a dose-dependent manner (Fig.

380

2E). Previous reports have showed that STING plays an important role in the

381

regulation of IFN-β induction in response to transfected poly(dA:dT) which is a

382

synthetic dsDNA that mimics dsDNA virus (21, 42). We verified that

383

poly(dA:dT)-induced IFN-β production is mostly dependent on STING by knocking

384

down STING expression in PH5CH8 cells which has the intact capacity for type I IFN

385

induction (43) (data not shown). Based on this, PH5CH8 cells expressing Pol or not

386

were used to further transfected with poly(dA:dT) to activate the STING signaling.

387

The results showed that less IFN-β mRNA was detected in cells expressing Pol

388

compared to that in control cells upon poly(dA:dT) stimulation (Fig.2F). In addition,

389

the impaired antiviral response mediated by STING was also observed in Huh7 cells

390

expressing Pol (Fig. 2G). 13 / 32

391

To examine whether Pol could act as an IFN- production blocker in the context of

392

HBV replication, we compared the response of Huh7 cells harbouring the wild type

393

pHBV1.3 or the polymerase-null mutant pHBV1.3-∆Pol upon cGAMP stimulation.

394

As shown in Fig.2G, in Huh7 cells transfected with the wild-type HBV, IFN-

395

promoter activation induced by cGAMP was obviously reduced, while not much

396

affected in those cells lacking HBV polymerase expression (Fig.2H). Similar results

397

were obtained in Huh7 cells transfected with either the wild-type pCMV-HBV

398

construct or the corresponding Pol-null mutant pCMV-HBV-∆Pol (data not shown),

399

thereby verifying the inhibitory effect of HBV Pol on the STING-mediated IFN-

400

production in the context of viral replication.

401

Considering Pol has an indispensible role in HBV genome replication as reverse

402

transcriptase, we intended to explore whether the Pol’s catalytic activity is involved in

403

its inhibitory effect on STING signaling. To test this point, we constructed the

404

polymerase-inactive mutant pHBV1.3-YMHD with which none of HBV DNA

405

replication intermediates wasdetected (data not shown) and tested its effect on

406

STING-mediated IFN-β production. We found that the mutated Pol exhibited the

407

comparable suppression effect on the IFN-β promoter activation with the wild type

408

Pol in Huh7 cells (Fig.2I, upper part). A similar observation was obtained with the

409

mutant pFlag-Pol-YMHD (Fig.2I, lower part). Thus, the results indicate that Pol

410

suppresses STING signaling in a manner that is independent of its catalytic activity.

411

Collectively, these results demonstrate that Pol is predominantly responsible for the

412

suppression of STING-mediated IFN- production by HBV in human hepatic cell

413

lines.

414

3. HBV polymerase physically associates with STING.

415

To gain further insight into how HBV polymerase disrupts STING signaling, we

416

determined whether Pol interacts with STING to inhibit its function. We first

417

examined if Pol and STING co-localize in Huh7 cells by immunofluorescence. As

418

shown in Fig. 3A (upper panel), Flag-tagged Pol exhibited a cytoplasmic punctate

419

pattern of immunofluorescence which moderately co-localized with STING, implying

420

that Pol may interact with STING. By contrast, the viral protein Core did not show 14 / 32

421

any co-localization with STING (Fig. 3A, lower panel). In view of these results, we

422

further confirmed the association of Pol with STING by co-immunoprecipitation

423

analysis. The Pol-expressing stable cell line 293T-Flag-Pol and the control cell line

424

293T-GFP were transfected with HA-tagged STING. The results showed that the

425

Flag-tagged Pol, but not the GFP control, could efficiently co-precipitate with the

426

STING protein (Fig. 3B), suggesting that Pol binds to STING. However, we did not

427

detect any interaction between Pol and the retinoic acid inducible gene (RIG-I), which

428

is known as an RNA sensor that can induce IFN-β production and also be inhibited by

429

Pol (10, 11) (Fig. 3C). These results suggested that the association between Pol and

430

STING is specific.

431

4. The RT/RH domain of Pol binds to STING and is responsible for the

432

inhibitory activity of Pol.

433

HBV polymerase is composed of four distinct regions, including the terminal protein

434

(TP), the spacer, the reverse transcriptase (RT), and the ribonuclease H (RNase H). To

435

delineate the region of Pol interacting with STING, a series of truncated mutants of

436

Pol were constructed (Fig. 4A). We co-transfected cells with the plasmids expressing

437

Flag-tagged truncated mutants of Pol together with HA-tagged STING and examined

438

the association between them by co-immunoprecipitation analysis. The results showed

439

that HA-tagged STING could bind to the full-length Pol, RT/RH, RT and RH

440

domains of Pol, but barely to the TP/Spacer region. Moreover, binding of RT/RH and

441

RT domains with STING exhibited high affinity (Fig. 4B, upper panel).

442

To test whether the association of Pol with STING resulted in the inhibition of

443

STING-mediated IFN- production, the truncated mutants of Pol were further

444

co-transfected with the STING-expressing and the IFN-β reporter plasmids into Huh7

445

cells. The results showed that the IFN-β promoter activation was greatly inhibited in

446

the cells expressing full-length Pol, RT/RH, RT and RH but was rarely affected in the

447

cells expressing TP and Spacer regions of Pol. The biggest inhibition of more than 80%

448

was exhibited by the RT/RH region of Pol (Fig. 4C), suggesting that the protein

449

sequence within the RT/RH region of Pol is critical to the inhibitory effect. A similar

450

effect of RT/RH domains on the IRF-regulating p55-C1B promoter activation was 15 / 32

451

shown in Fig. 4D. To further test the inhibitory effect of the RT and RH domains on

452

IFN- production, we conducted the dose response assay using different amounts of

453

the RT and RH mutants. The results demonstrated that just 100ng of the Pol RT

454

domain could decrease the IFN-β promoter activation by almost 70%, meanwhile, the

455

RH mutant also had an inhibitory effect on the IFN- promoter activation albeit at a

456

milder rate (Fig. 4E). Additionally, overexpression of the mutant ∆RH reduced the

457

IFN-β promoter activation by approximately 50% compared to the control, by

458

contrast, ∆RT mutant only modestly downregulated the IFN-β promoter activation

459

(Fig. 4C). Thus, the RT domain of HBV polymerase relatively makes more

460

contribution on the inhibition of STING-mediated IFN-β induction by Pol, while the

461

RH domain is subordinately responsible for the inhibitory effect. These could be

462

combined with the above data to suggest that the binding of Pol to STING associated

463

with its inhibitory activity on interferon induction, and the binding affinity positively

464

correlated with the degree of the inhibitory effect. We thus concluded that Pol may

465

interact with STING through the RT and RH domains and consequently suppresses

466

STING-mediated IFN- induction in the transfection system.

467

5. The K63-linked polyubiquitination of STING is drastically decreased by HBV

468

polymerase.

469

Modification of signaling molecules by ubiquitination plays a central role in

470

transmitting signals important for activation of innate antiviral responses (44). Recent

471

studies indicate that STING polyubiquitination is required for the activation of IFN

472

induction pathway (45, 46). Ubiquitination events structurally contain several distinct

473

ubiquitin chain linkage types (47). The best characterized linkages to date are the K48

474

ubiquitin chain and the K63 ubiquitin chain (48). K48-linked polyubiquitin mainly

475

targets proteins for proteasomal degradation, whereas K63-linked polyubiquitin more

476

often regulates protein function, subcellular localization, or protein-protein interaction.

477

In view of our above observations and to define how the interaction between Pol and

478

STING abrogates the STING-mediated IFN-β production, we first determined if Pol

479

impairs STING polyubiquitination by in vivo ubiquitination analysis in HEK293T

480

cells. A dramatic decrease in STING ubiquitination was observed in cells expressing 16 / 32

481

Pol compared to that in control cells (Fig. 5A). Meanwhile, the expression levels of

482

STING in the presence and absence of HBV Pol expression remained comparable,

483

suggesting that the linkage of STING ubiquitination impaired by Pol is most likely

484

K63 (Fig. 1 B and Fig. 5A, middle panels). To confirm this point, we employed two

485

plasmids including pHA-Ub-K48R, in which the lysine at position 48 of the ubiquitin

486

was substituted with arginine, and pHA-Ub-K63, in which all lysines except K63

487

were mutated to arginines. The cells transfected with HA-Ub-K48R still showed a

488

reduction in the level of ubiquitinated STING in the presence of Pol (Fig. 5B).

489

Moreover, the K63-linked ubiquitination of STING which has been shown to be

490

important for cellular antiviral response (45, 46), was markedly decreased in the cells

491

expressing Pol, indicating that HBV Pol inhibits IFN- production by specifically

492

targeting the K63- rather than the K48-linked ubiquitination of STING (Fig. 5C).

493

Subsequently, we confirmed this in the context of HBV replication using pHBV1.3 or

494

pHBV1.3-∆Pol in Huh7 cells and found that the K63-linked ubiquitination of STING

495

was decreased in the cells transfected with the wild type HBV genome, however, this

496

attenuation was rescued in the cells containing the HBV Pol-null mutant (Fig. 5D).

497

Next, to define which domain(s) of Pol contribute to the disruption of STING

498

ubiquitination, we co-transfected the series of Pol truncated mutants together with

499

STING and HA-Ub-K63 plasmids followed by examination of STING ubiquitination.

500

Expression of truncated mutants of Pol downregulated K63-linked ubiquitination of

501

STING to a different extent, with the biggest decrease exhibited by the full length Pol,

502

RT/RH domain and RT domain, followed by ∆RT and ∆RH mutants and a weak

503

decrease exhibited by TP/Spacer and RH domains (Fig. 5E, top panel). These results

504

were largely in agreement with our previous observations on their inhibitory effects

505

on STING signaling (Fig. 4B-4E) and suggest that the impairment of K63-linked

506

ubiquitination of STING by Pol may depend on the binding of Pol to STING, thereby

507

leading to a weakened IFN-β production and antiviral response.

508

6. HBV polymerase did not affect the expression of the E3ubiquitin ligases

509

TRIM32 and TRIM56 and their interaction with STING.

510

The E3ubiquitin ligases tripartite motif protein 32 (TRIM32) and TRIM56 have been 17 / 32

511

identified to target STING for K63-linked ubiquitination (45, 46). Thus we speculate

512

whether Pol disrupts the K63-linked ubiquitination of STING by competitively

513

binding to STING that associates with these two E3 ligases. To this end, we examined

514

the impact of Pol on the interaction of TRIM32 or TRIM56 with STING by

515

co-immunoprecipitation analysis. The results showed that the expression levels of

516

TRIM32 and TRIM56 were comparable, regardless of the presence or absence of Pol

517

(Fig.6A, input part). Moreover, the interaction between STING and TRIM32 or

518

TRIM56 was not apparently affected by Pol in the overexpression system (Fig. 6A,

519

top panel).To further confirm this result, we pulled down the endogenous TRIM32 or

520

TRIM56 by Flag-tagged STING with or without Pol. Analysis of the

521

immunoprecipitated products showed that Pol had little effect on the binding of

522

STING to endogenous TRIM32 (Fig. 6B). However, we failed to detect the

523

interaction between STING and endogenous TRIM56 in 293 cells, which may be due

524

to the expression of TRIM56 in the cells is rather low (data not shown). Taken

525

together, these data suggested the suppression of STING K63-linked ubiquitination by

526

Pol was not mediated by a disturbance of the expression of TRIM32 or TRIM56 and

527

their interaction with STING.

528 529

Discussion

530

It has long been controversial whether HBV induces innate immune responses during

531

its infection. A previous study using the chimpanzee model showed that HBV didn’t

532

activate the host innate antiviral responses in the liver, and thus it was described as a

533

‘stealth virus’ (49, 50). Nevertheless, several recent observations in clinic specimens,

534

chimeric mouse models and infectious cell models have shown that HBV may induce

535

some type I IFN and type III IFN production, suggesting that the host can sense the

536

HBV infection and then initiates the innate immune responses (51-55). A possible

537

explanation for the conflicts is that HBV has evolved numerous strategies to block the

538

host antiviral responses early in infection (6-11, 56-59) and thus it is hard for the

539

investigators to detect the host innate antiviral responses during its infection. In the

540

present study, we found that HBV could escape host innate immune response via 18 / 32

541

antagonizing IFN-β induction mediated by STING, a key molecule in the host

542

cytosolic DNA-sensing pathways (17, 18, 20, 38, 60). Our results revealed, for the

543

first time, that the HBV polymerase interacts with STING and inhibits its K63-linked

544

ubiquitination associated with loss of STING function and consequent impairment of

545

IRF3 activation and IFN-β induction and antiviral response.

546

Given the key role of STING in the regulation of host antiviral response, many

547

viruses have been reported to target it for subversion of the host innate immunity by

548

various elaborate mechanisms. It has been shown that HCV, also a hepatotropic virus

549

with a high rate of persistence, inhibits STING-mediated signaling through the viral

550

NS4B protein (40, 61). The protease NS2B3 of dengue virus can cleave STING to

551

block the type I IFN induction (62, 63), and the papain-like proteases of coronaviruses

552

interfere with the STING-mediated signaling pathway by acting as viral

553

deubiquitinases (64, 65). For HBV, though the PRRs and related signaling pathways

554

involved in sensing HBV infection and triggering antiviral innate immune responses

555

remain to be clarified clearly, many reports have shown that activation of the PRR

556

pathways specifically inhibit the HBV replication (6, 26, 54, 66, 67). Moreover, DAI,

557

a cytosolic DNA sensor that also utilizes STING as an adaptor (68), was also showed

558

to inhibit HBV replication in our previous work (69), implying that suppression of the

559

activation of STING, the converging point of the PRRs signaling, may be an efficient

560

strategy for HBV, like many other viruses, to evade the innate antiviral activity at an

561

early stage.

562

Although several cellular biological events, such as STING ubiquitination,

563

dimerization and its recruitment of TANK-binding kinase 1 (TBK1) (41, 70), are

564

reported to be critical for STING-mediated signal transduction, more investigations

565

are needed to characterize the detailed mechanisms that control STING activation and

566

signaling (71). In this study, we demonstrated that HBV Pol markedly impaired

567

K63-linked ubiquitination of STING by physically associating with it through the

568

RT/RH region, though had moderate effect on STING dimerization, which is essential

569

for STING self-activation and subsequent downstream signaling culminating in the

570

induction of type I IFN (21) (data not shown). The recruitment of TBK1 and IRF3 by 19 / 32

571

STING is also required for the activation of STING signaling and appears to be a

572

process that regulated by STING ubiquitin modification. Although K63-linked

573

ubiquitination of STING was significantly inhibited by Pol, the results obtained in

574

293T cells didn’t show that Pol could affect the interaction between overexpressed

575

TBK1 and STING (data not shown). Since the K63-ubiquitination of STING is not

576

only important for STING to recruit TBK1/IRF3, but also critical for STING as a

577

scaffold protein to promote TBK1 and IRF3 activation, the observations we’ve found

578

that Pol-mediated disruption of K63-linked ubiquitination of STING didn’t affect the

579

recruitment of TBK1 to STING but resulted in the significant inhibition of IRF3

580

phosphorylation (data not shown) and dimerization and IFN induction were

581

reasonable. Additionally, while the essential role of STING in DNA sensing pathways

582

is well-established, it has also been shown to play a role in RIG-I-mediated IFN-β

583

production (22, 24). Furthermore, HBV Pol has been reported to inhibit

584

RIG-I-mediated signaling by disrupting the interaction between TBK1/IKKε and

585

DDX3 (10, 11). Therefore, there is a possibility that the suppression effect of Pol on

586

RIG-I signaling is involved in its antagonism on STING-mediated IFN-β induction.

587

To test this hypothesis, we examined whether overexpression of DDX3 could restore

588

the inhibitory effect of HBV polymerase on STING signaling and found that the

589

suppressed IFN-β promoter activity could be efficiently rescued by over-expressed

590

DDX3 when RIG-I but not STING acted as the stimulator (data not shown). This

591

result partly exclude the possibility that Pol inhibits STING signaling to a large extent

592

through its act on RIG-I signaling. Therefore, we concluded that HBV Pol inhibits

593

STING-mediated type I IFN production mainly by dampening its K63-linked

594

ubiquitination. Nevertheless, the precise delineation how Pol affects STING-mediated

595

signaling requires further understanding of the molecular mechanisms involved in

596

STING activation.

597

HBV Pol consists of four domains. The RT domain is responsible for reverse

598

transcription and nucleocapsid assembly of HBV (72-74), while the TP domain serves

599

as a protein primer to initiate reverse transcription (75). The C-terminal RNase H

600

domain directly mediates the degradation of pregenomic RNA template after reverse 20 / 32

601

transcription to maintain the stability of HBV genome. It should be noted that Pol is

602

commonly believed to be inefficiently translated and rapidly encapsidated. However,

603

some investigators detected non-encapsidated polymerase accumulated in the

604

cytoplasm in a manner similar to non-encapsidated duck hepatitis B virus (DHBV)

605

polymerase (76, 77), which implies that Pol may have additional functions besides

606

reverse transcriptase. Recent reports implicate Pol displays immunomodulatory

607

activities through binding to a series of host factors (78). Besides blocking RIG-I- and

608

TLR3-mediated IFN-β induction via dampening the interaction between TBK1/IKKε

609

and DDX3 as mentioned above, Pol was also shown to abrogate IFN-α signaling by

610

suppressing the nuclear translocation of STAT1 and STAT2 in Huh7 cells (79).

611

Interestingly, the detailed studies of the TP domain of Pol revealed its association with

612

PKC-δ resulting in inhibition of PKC-δ phosphorylation, meanwhile, RH domain was

613

able to interfere with nuclear transportation of STAT1/2 through a competitive binding

614

to importin-α5 (80). Here, we demonstrated that the RT and RH domains of Pol were

615

responsible for the inhibition of STING-activated signaling transduction through

616

associating with STING and inhibiting its activation. Overall, these investigations

617

support a notion that besides its inherent catalytic role in viral replication, Pol has

618

multifunctional activities, including regulating host immune signaling pathways by

619

binding to diverse host targets, which may contribute to HBV evasion of the innate

620

immunity. Nevertheless, further work should aim to determine the significance of

621

Pol-mediated immune modulation during HBV infection at more physiological levels

622

of Pol expression in vivo.

623

In summary, a new role of HBV polymerase in inhibition of IFN-β production

624

through selectively targeting the cytosolic DNA sensor and the adaptor molecule

625

STING has been proposed here. We believe that these findings may promote our

626

understanding on the molecular mechanisms by which HBV escapes the innate

627

immunity and establishes chronic infection and provide new target for designing

628

novel therapeutic interventions to combat HBV.

629 630

Acknowledgments 21 / 32

631

We thank Dr. Takashi Fujita and Dr. Hiroki Kato (Institute for Virus Research, Kyoto

632

University, Kyoto, Japan) for the insightful suggestions and kindly providing the

633

plasmids (pEF-STING, pEF-STING-Myc, and p-55C1B-Luc). We also thank Dr.

634

Hongbing Shu (The College of Life Sciences, Wuhan University, Wuhan, China) for

635

his generous gift of the plasmids (pHA-MITA, pFlag-MITA, pFlag-TRIM32,

636

pFlag-TRIM56 and the RNAi plasmids targeting human STING) and the

637

anti-TRIM32 antibody. We are indebted to Dr. Shiyan Yu for good advice and full

638

support. We are grateful to Dr. Zekun Wang for preparing several of the truncations

639

of Pol and the ubiquitin mutants pHA-Ub-K48R and pHA-Ub-K63 used in this study

640

and helpful discussions. We thank Dr. Maya Kozlowski for critical review of the

641

manuscript.

642

This research was supported by the National Key Basic Research Program of China

643

(2012CB519000 to Z.Y.), the National Megaprojects of China for infectious Diseases

644

(2012ZX10002007-001 to Z.Y.), the German Research Foundation (SFB/Transregio

645

TRR60 to Z.Y.), the National Natural Science Foundation of China (31200129 to J.

646

Li), the China Postdoctoral Science Foundation (201104232 to J. Li), and the China

647

Postdoctoral Science Foundation (2014M551325 to J.L. Chen).

648 649

References

650 651 652 653 654 655 656 657 658 659 660 661 662 663 664

1.

Dienstag, J. L. 2008. Hepatitis B virus infection. The New England journal of medicine 359:1486-1500.

2.

Gonzalez, S. A., and E. B. Keeffe. 2011. Chronic viral hepatitis: epidemiology, molecular biology, and antiviral therapy. Frontiers in bioscience 16:225-250.

3.

Ait-Goughoulte, M., J. Lucifora, F. Zoulim, and D. Durantel. 2010. Innate antiviral immune responses to hepatitis B virus. Viruses 2:1394-1410.

4.

Cunha, A. 2012. Innate immunity: Pathogen and xenobiotic sensing - back to basics. Nature reviews. Immunology 12:400.

5.

Ward, C. J. 2010. Pathogen sensing in innate immunity. Expert review of vaccines 9:19-21.

6.

Wu, J., Z. Meng, M. Jiang, R. Pei, M. Trippler, R. Broering, A. Bucchi, J. P. Sowa, U. Dittmer, D. Yang, M. Roggendorf, G. Gerken, M. Lu, and J. F. Schlaak. 2009. Hepatitis B virus suppresses toll-like receptor-mediated innate immune responses in murine parenchymal and nonparenchymal liver cells. Hepatology 49:1132-1140.

7.

Kumar, M., S. Y. Jung, A. J. Hodgson, C. R. Madden, J. Qin, and B. L. Slagle. 2011. Hepatitis B virus regulatory HBx protein binds to adaptor protein IPS-1 and inhibits the 22 / 32

665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708

activation of beta interferon. Journal of virology 85:987-995. 8.

Wei, C., C. Ni, T. Song, Y. Liu, X. Yang, Z. Zheng, Y. Jia, Y. Yuan, K. Guan, Y. Xu, X. Cheng, Y. Zhang, X. Yang, Y. Wang, C. Wen, Q. Wu, W. Shi, and H. Zhong. 2010. The hepatitis B virus X protein disrupts innate immunity by downregulating mitochondrial antiviral signaling protein. Journal of immunology 185:1158-1168.

9.

Jiang, J., and H. Tang. 2010. Mechanism of inhibiting type I interferon induction by hepatitis B virus X protein. Protein & cell 1:1106-1117.

10.

Yu, S., J. Chen, M. Wu, H. Chen, N. Kato, and Z. Yuan. 2010. Hepatitis B virus polymerase inhibits RIG-I- and Toll-like receptor 3-mediated beta interferon induction in human hepatocytes through interference with interferon regulatory factor 3 activation and dampening of the interaction between TBK1/IKKepsilon and DDX3. The Journal of general virology 91:2080-2090.

11.

Wang, H., and W. S. Ryu. 2010. Hepatitis B virus polymerase blocks pattern recognition receptor signaling via interaction with DDX3: implications for immune evasion. PLoS pathogens 6:e1000986.

12.

Wang, Z., M. K. Choi, T. Ban, H. Yanai, H. Negishi, Y. Lu, T. Tamura, A. Takaoka, K. Nishikura, and T. Taniguchi. 2008. Regulation of innate immune responses by DAI (DLM-1/ZBP1) and other DNA-sensing molecules. Proceedings of the National Academy of Sciences of the United States of America 105:5477-5482.

13.

Takaoka, A., Z. Wang, M. K. Choi, H. Yanai, H. Negishi, T. Ban, Y. Lu, M. Miyagishi, T. Kodama, K. Honda, Y. Ohba, and T. Taniguchi. 2007. DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature 448:501-505.

14.

Rathinam, V. A., Z. Jiang, S. N. Waggoner, S. Sharma, L. E. Cole, L. Waggoner, S. K. Vanaja, B. G. Monks, S. Ganesan, E. Latz, V. Hornung, S. N. Vogel, E. Szomolanyi-Tsuda, and K. A. Fitzgerald. 2010. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nature immunology 11:395-402.

15.

Hornung, V., A. Ablasser, M. Charrel-Dennis, F. Bauernfeind, G. Horvath, D. R. Caffrey, E. Latz, and K. A. Fitzgerald. 2009. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature 458:514-518.

16.

Fernandes-Alnemri, T., J. W. Yu, P. Datta, J. Wu, and E. S. Alnemri. 2009. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 458:509-513.

17.

Unterholzner, L., S. E. Keating, M. Baran, K. A. Horan, S. B. Jensen, S. Sharma, C. M. Sirois, T. Jin, E. Latz, T. S. Xiao, K. A. Fitzgerald, S. R. Paludan, and A. G. Bowie. 2010. IFI16 is an innate immune sensor for intracellular DNA. Nature immunology 11:997-1004.

18.

Zhang, Z., B. Yuan, M. Bao, N. Lu, T. Kim, and Y. J. Liu. 2011. The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells. Nature immunology 12:959-965.

19.

Wu, J., L. Sun, X. Chen, F. Du, H. Shi, C. Chen, and Z. J. Chen. 2013. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science 339:826-830.

20.

Sun, L., J. Wu, F. Du, X. Chen, and Z. J. Chen. 2013. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339:786-791.

21.

Sun, W., Y. Li, L. Chen, H. Chen, F. You, X. Zhou, Y. Zhou, Z. Zhai, D. Chen, and Z. Jiang. 2009. ERIS, an endoplasmic reticulum IFN stimulator, activates innate immune 23 / 32

709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752

signaling through dimerization. Proceedings of the National Academy of Sciences of the United States of America 106:8653-8658. 22.

Zhong, B., Y. Yang, S. Li, Y. Y. Wang, Y. Li, F. Diao, C. Lei, X. He, L. Zhang, P. Tien, and H. B. Shu. 2008. The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity 29:538-550.

23.

Jin, L., P. M. Waterman, K. R. Jonscher, C. M. Short, N. A. Reisdorph, and J. C. Cambier. 2008. MPYS, a novel membrane tetraspanner, is associated with major histocompatibility complex class II and mediates transduction of apoptotic signals. Molecular and cellular biology 28:5014-5026.

24.

Ishikawa, H., and G. N. Barber. 2008. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 455:674-678.

25.

Burdette, D. L., K. M. Monroe, K. Sotelo-Troha, J. S. Iwig, B. Eckert, M. Hyodo, Y. Hayakawa, and R. E. Vance. 2011. STING is a direct innate immune sensor of cyclic di-GMP. Nature 478:515-518.

26.

Chen, J., and Z. Yuan. 2014. Interplay between hepatitis B virus and the innate immune responses: implications for new therapeutic strategies. Virologica Sinica 29:17-24.

27.

Wang, Z., J. Ni, J. Li, B. Shi, Y. Xu, and Z. Yuan. 2011. Inhibition of hepatitis B virus replication by cIAP2 involves accelerating the ubiquitin-proteasome-mediated destruction of polymerase. Journal of virology 85:11457-11467.

28.

Li, J., Y. Liu, Z. Wang, K. Liu, Y. Wang, J. Liu, H. Ding, and Z. Yuan. 2011. Subversion of cellular autophagy machinery by hepatitis B virus for viral envelopment. Journal of virology 85:6319-6333.

29.

Li, J., S. Lin, Q. Chen, L. Peng, J. Zhai, Y. Liu, and Z. Yuan. 2010. Inhibition of hepatitis B virus replication by MyD88 involves accelerated degradation of pregenomic RNA and nuclear retention of pre-S/S RNAs. Journal of virology 84:6387-6399.

30.

Gao, W., and J. Hu. 2007. Formation of hepatitis B virus covalently closed circular DNA: removal of genome-linked protein. Journal of virology 81:6164-6174.

31.

Jones, S. A., R. Boregowda, T. E. Spratt, and J. Hu. 2012. In vitro epsilon RNA-dependent protein priming activity of human hepatitis B virus polymerase. Journal of virology 86:5134-5150.

32.

Ladner, S. K., M. J. Otto, C. S. Barker, K. Zaifert, G. H. Wang, J. T. Guo, C. Seeger, and R. W. King. 1997. Inducible expression of human hepatitis B virus (HBV) in stably transfected hepatoblastoma cells: a novel system for screening potential inhibitors of HBV replication. Antimicrobial agents and chemotherapy 41:1715-1720.

33.

Noguchi, M., and S. Hirohashi. 1996. Cell lines from non-neoplastic liver and hepatocellular carcinoma tissue from a single patient. In vitro cellular & developmental biology. Animal 32:135-137.

34.

Gripon, P., S. Rumin, S. Urban, J. Le Seyec, D. Glaise, I. Cannie, C. Guyomard, J. Lucas, C. Trepo, and C. Guguen-Guillouzo. 2002. Infection of a human hepatoma cell line by hepatitis B virus. Proceedings of the National Academy of Sciences of the United States of America 99:15655-15660.

35.

Yan, H., G. Zhong, G. Xu, W. He, Z. Jing, Z. Gao, Y. Huang, Y. Qi, B. Peng, H. Wang, L. Fu, M. Song, P. Chen, W. Gao, B. Ren, Y. Sun, T. Cai, X. Feng, J. Sui, and W. Li. 2012. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B 24 / 32

753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796

and D virus. eLife 1:e00049. 36.

Woodward, J. J., A. T. Iavarone, and D. A. Portnoy. 2010. c-di-AMP secreted by intracellular Listeria monocytogenes activates a host type I interferon response. Science 328:1703-1705.

37.

Gripon, P., C. Diot, N. Theze, I. Fourel, O. Loreal, C. Brechot, and C. Guguen-Guillouzo. 1988. Hepatitis B virus infection of adult human hepatocytes cultured in the presence of dimethyl sulfoxide. Journal of virology 62:4136-4143.

38.

Diner, E. J., D. L. Burdette, S. C. Wilson, K. M. Monroe, C. A. Kellenberger, M. Hyodo, Y. Hayakawa, M. C. Hammond, and R. E. Vance. 2013. The innate immune DNA sensor cGAS produces a noncanonical cyclic dinucleotide that activates human STING. Cell reports 3:1355-1361.

39.

Zhang, X., H. Shi, J. Wu, X. Zhang, L. Sun, C. Chen, and Z. J. Chen. 2013. Cyclic GMP-AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING. Molecular cell 51:226-235.

40.

Ding, Q., X. Cao, J. Lu, B. Huang, Y. J. Liu, N. Kato, H. B. Shu, and J. Zhong. 2013. Hepatitis C virus NS4B blocks the interaction of STING and TBK1 to evade host innate immunity. Journal of hepatology 59:52-58.

41.

Tanaka, Y., and Z. J. Chen. 2012. STING specifies IRF3 phosphorylation by TBK1 in the cytosolic DNA signaling pathway. Science signaling 5:ra20.

42.

Ishikawa, H., Z. Ma, and G. N. Barber. 2009. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 461:788-792.

43.

Li, K., Z. Chen, N. Kato, M. Gale, Jr., and S. M. Lemon. 2005. Distinct poly(I-C) and virus-activated signaling pathways leading to interferon-beta production in hepatocytes. The Journal of biological chemistry 280:16739-16747.

44.

Bhoj, V. G., and Z. J. Chen. 2009. Ubiquitylation in innate and adaptive immunity. Nature 458:430-437.

45.

Zhang, J., M. M. Hu, Y. Y. Wang, and H. B. Shu. 2012. TRIM32 protein modulates type I interferon induction and cellular antiviral response by targeting MITA/STING protein for K63-linked ubiquitination. The Journal of biological chemistry 287:28646-28655.

46.

Tsuchida, T., J. Zou, T. Saitoh, H. Kumar, T. Abe, Y. Matsuura, T. Kawai, and S. Akira. 2010. The ubiquitin ligase TRIM56 regulates innate immune responses to intracellular double-stranded DNA. Immunity 33:765-776.

47.

Malynn, B. A., and A. Ma. 2010. Ubiquitin makes its mark on immune regulation. Immunity 33:843-852.

48.

Newton, K., M. L. Matsumoto, I. E. Wertz, D. S. Kirkpatrick, J. R. Lill, J. Tan, D. Dugger, N. Gordon, S. S. Sidhu, F. A. Fellouse, L. Komuves, D. M. French, R. E. Ferrando, C. Lam, D. Compaan, C. Yu, I. Bosanac, S. G. Hymowitz, R. F. Kelley, and V. M. Dixit. 2008. Ubiquitin chain editing revealed by polyubiquitin linkage-specific antibodies. Cell 134:668-678.

49.

Wieland, S., R. Thimme, R. H. Purcell, and F. V. Chisari. 2004. Genomic analysis of the host response to hepatitis B virus infection. Proceedings of the National Academy of Sciences of the United States of America 101:6669-6674.

50.

Wieland, S. F., and F. V. Chisari. 2005. Stealth and cunning: hepatitis B and hepatitis C viruses. Journal of virology 79:9369-9380. 25 / 32

797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840

51.

Fisicaro, P., C. Valdatta, C. Boni, M. Massari, C. Mori, A. Zerbini, A. Orlandini, L. Sacchelli, G. Missale, and C. Ferrari. 2009. Early kinetics of innate and adaptive immune responses during hepatitis B virus infection. Gut 58:974-982.

52.

Hosel, M., M. Quasdorff, K. Wiegmann, D. Webb, U. Zedler, M. Broxtermann, R. Tedjokusumo, K. Esser, S. Arzberger, C. J. Kirschning, A. Langenkamp, C. Falk, H. Buning, S. Rose-John, and U. Protzer. 2009. Not interferon, but interleukin-6 controls early gene expression in hepatitis B virus infection. Hepatology 50:1773-1782.

53.

Lucifora, J., D. Durantel, B. Testoni, O. Hantz, M. Levrero, and F. Zoulim. 2010. Control of hepatitis B virus replication by innate response of HepaRG cells. Hepatology 51:63-72.

54.

Lutgehetmann, M., T. Bornscheuer, T. Volz, L. Allweiss, J. H. Bockmann, J. M. Pollok, A. W. Lohse, J. Petersen, and M. Dandri. 2011. Hepatitis B virus limits response of human hepatocytes to interferon-alpha in chimeric mice. Gastroenterology 140:2074-2083, 2083 e2071-2072.

55.

Shlomai, A., R. E. Schwartz, V. Ramanan, A. Bhatta, Y. P. de Jong, S. N. Bhatia, and C. M. Rice. 2014. Modeling host interactions with hepatitis B virus using primary and induced pluripotent stem cell-derived hepatocellular systems. Proceedings of the National Academy of Sciences of the United States of America 111:12193-12198.

56.

Christen, V., F. Duong, C. Bernsmeier, D. Sun, M. Nassal, and M. H. Heim. 2007. Inhibition of alpha interferon signaling by hepatitis B virus. Journal of virology 81:159-165.

57.

Visvanathan, K., N. A. Skinner, A. J. Thompson, S. M. Riordan, V. Sozzi, R. Edwards, S. Rodgers, J. Kurtovic, J. Chang, S. Lewin, P. Desmond, and S. Locarnini. 2007. Regulation of Toll-like receptor-2 expression in chronic hepatitis B by the precore protein. Hepatology 45:102-110.

58.

Chen, Z., Y. Cheng, Y. Xu, J. Liao, X. Zhang, Y. Hu, Q. Zhang, J. Wang, Z. Zhang, F. Shen, and Z. Yuan. 2008. Expression profiles and function of Toll-like receptors 2 and 4 in peripheral blood mononuclear cells of chronic hepatitis B patients. Clinical immunology 128:400-408.

59.

Oliviero, B., S. Varchetta, E. Paudice, G. Michelone, M. Zaramella, D. Mavilio, F. De Filippi, S. Bruno, and M. U. Mondelli. 2009. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology 137:1151-1160, 1160 e1151-1157.

60.

Abe, T., A. Harashima, T. Xia, H. Konno, K. Konno, A. Morales, J. Ahn, D. Gutman, and G. N. Barber. 2013. STING recognition of cytoplasmic DNA instigates cellular defense. Molecular cell 50:5-15.

61.

Nitta, S., N. Sakamoto, M. Nakagawa, S. Kakinuma, K. Mishima, A. Kusano-Kitazume, K. Kiyohashi, M. Murakawa, Y. Nishimura-Sakurai, S. Azuma, M. Tasaka-Fujita, Y. Asahina, M. Yoneyama, T. Fujita, and M. Watanabe. 2013. Hepatitis C virus NS4B protein targets STING and abrogates RIG-I-mediated type I interferon-dependent innate immunity. Hepatology 57:46-58.

62.

Aguirre, S., A. M. Maestre, S. Pagni, J. R. Patel, T. Savage, D. Gutman, K. Maringer, D. Bernal-Rubio, R. S. Shabman, V. Simon, J. R. Rodriguez-Madoz, L. C. Mulder, G. N. Barber, and A. Fernandez-Sesma. 2012. DENV inhibits type I IFN production in infected cells by cleaving human STING. PLoS pathogens 8:e1002934.

63.

Yu, C. Y., T. H. Chang, J. J. Liang, R. L. Chiang, Y. L. Lee, C. L. Liao, and Y. L. Lin. 26 / 32

841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884

2012. Dengue virus targets the adaptor protein MITA to subvert host innate immunity. PLoS pathogens 8:e1002780. 64.

Sun, L., Y. Xing, X. Chen, Y. Zheng, Y. Yang, D. B. Nichols, M. A. Clementz, B. S. Banach, K. Li, S. C. Baker, and Z. Chen. 2012. Coronavirus papain-like proteases negatively regulate antiviral innate immune response through disruption of STING-mediated signaling. PloS one 7:e30802.

65.

Xing, Y., J. Chen, J. Tu, B. Zhang, X. Chen, H. Shi, S. C. Baker, L. Feng, and Z. Chen. 2013. The papain-like protease of porcine epidemic diarrhea virus negatively regulates type I interferon pathway by acting as a viral deubiquitinase. The Journal of general virology 94:1554-1567.

66.

Wu, J., S. Huang, X. Zhao, M. Chen, Y. Lin, Y. Xia, C. Sun, X. Yang, J. Wang, Y. Guo, J. Song, E. Zhang, B. Wang, X. Zheng, J. F. Schlaak, M. Lu, and D. Yang. 2014. Poly(I:C) treatment leads to interferon-dependent clearance of hepatitis B virus in a hydrodynamic injection mouse model. Journal of virology 88:10421-10431.

67.

Han, Q., C. Zhang, J. Zhang, and Z. Tian. 2011. Reversal of hepatitis B virus-induced immune tolerance by an immunostimulatory 3p-HBx-siRNAs in a retinoic acid inducible gene I-dependent manner. Hepatology 54:1179-1189.

68.

DeFilippis, V. R., D. Alvarado, T. Sali, S. Rothenburg, and K. Fruh. 2010. Human cytomegalovirus induces the interferon response via the DNA sensor ZBP1. Journal of virology 84:585-598.

69.

Chen, Q. Y., Y. H. Liu, J. H. Li, Z. K. Wang, J. X. Liu, and Z. H. Yuan. 2012. DNA-dependent activator of interferon-regulatory factors inhibits hepatitis B virus replication. World journal of gastroenterology : WJG 18:2850-2858.

70.

Shu, C., G. Yi, T. Watts, C. C. Kao, and P. Li. 2012. Structure of STING bound to cyclic di-GMP reveals the mechanism of cyclic dinucleotide recognition by the immune system. Nature structural & molecular biology 19:722-724.

71.

Burdette, D. L., and R. E. Vance. 2013. STING and the innate immune response to nucleic acids in the cytosol. Nature immunology 14:19-26.

72.

Summers, J., and W. S. Mason. 1982. Replication of the genome of a hepatitis B--like virus by reverse transcription of an RNA intermediate. Cell 29:403-415.

73.

Hirsch, R. C., J. E. Lavine, L. J. Chang, H. E. Varmus, and D. Ganem. 1990. Polymerase gene products of hepatitis B viruses are required for genomic RNA packaging as wel as for reverse transcription. Nature 344:552-555.

74.

Bartenschlager, R., and H. Schaller. 1992. Hepadnaviral assembly is initiated by polymerase binding to the encapsidation signal in the viral RNA genome. The EMBO journal 11:3413-3420.

75.

Pollack, J. R., and D. Ganem. 1993. An RNA stem-loop structure directs hepatitis B virus genomic RNA encapsidation. Journal of virology 67:3254-3263.

76.

Cao, F., and J. E. Tavis. 2004. Detection and characterization of cytoplasmic hepatitis B virus reverse transcriptase. The Journal of general virology 85:3353-3360.

77.

Yao, E., and J. E. Tavis. 2003. Kinetics of synthesis and turnover of the duck hepatitis B virus reverse transcriptase. The Journal of biological chemistry 278:1201-1205.

78.

Jones, S. A., and J. Hu. 2013. Hepatitis B virus reverse transcriptase: diverse functions as classical and emerging targets for antiviral intervention. Emerging Microbes and Infections 27 / 32

885 886 887 888 889 890 891 892 893

2:e56. 79.

Wu, M., Y. Xu, S. Lin, X. Zhang, L. Xiang, and Z. Yuan. 2007. Hepatitis B virus polymerase inhibits the interferon-inducible MyD88 promoter by blocking nuclear translocation of Stat1. The Journal of general virology 88:3260-3269.

80.

Chen, J., M. Wu, X. Zhang, W. Zhang, Z. Zhang, L. Chen, J. He, Y. Zheng, C. Chen, F. Wang, Y. Hu, X. Zhou, C. Wang, Y. Xu, M. Lu, and Z. Yuan. 2013. Hepatitis B virus polymerase impairs interferon-alpha-induced STA T activation through inhibition of importin-alpha5 and protein kinase C-delta. Hepatology 57:470-482.

894

Figure legends

895

Fig.1 HBV inhibits STING-mediated IFN-β induction and antiviral response.

896

(A, B) Huh7 cells were co-transfected with pIFN-β-Luc (A) or p-55C1B-Luc (B),

897

pRL-TK and 100ng, 200ng, 400ng of the plasmids pHBV1.3 or pCMV-HBV together

898

with pSTING (80ng) or empty vector. After 36h, the cells were extracted and assayed

899

for luciferase activity. The expression of STING in cells was examined by Western

900

blot and the expression of β-actin was examined as a loading control. Meanwhile, the

901

level of core capsids was detected using anti-Core antibody as described in Materials

902

and Methods (A, right part). (C) Measurement of endogeous STING expression in the

903

indicated cell lines by Western blot with anti-STING antibodies. (D) Huh7 cells were

904

co-transfected with pIFN-β-Luc, pRL-TK, and 125ng, 250ng, 500ng of the plasmids

905

pHBV1.3 or pCMV-HBV together with pSTING (25ng) or empty vector. After 36h,

906

the cells were mock-treated or treated with the indicated concentration of cGAMP for

907

additional 16h and then harvested for luciferase assay. Data are expressed as the mean

908

fold induction ± SD relative to control levels. (E, F) Huh7 cells were co-transfected

909

with 125ng, 250ng, 500ng of pHBV1.3 together with pSTING (25ng) or empty vector.

910

After 36h, the cells were mock-treated or treated with 100nM cGAMP for 16h. The

911

amount of IFN-β in the supernatants was determined by ELISA (E). Meanwhile, the

912

remaining supernatants from Huh7 cells were then collected and transferred to Vero

913

cells for overnight incubation with a positive control using IFN-a (500 IU ml-1)

914

treatment. Vero cells were then infected with NDV-GFP (80 HAU ml-1) for additional

915

24h. The GFP expression levels were determined by microscopy observation and

916

Western blot (F). (G) HepAD38 cells cultured in the presence or absence of

917

doxcycline were co-transfected with pIFN-β-Luc, pRL-TK and pSTING (25ng) or 28 / 32

918

empty vector. After 36h, the cells were mock-treated or treated with the indicated

919

concentration of cGAMP for additional 16h and then harvested for luciferase assay.

920

(H) HepaRG cells were seeded onto the chambered coverglass and proliferated and

921

differentiated. At day 10 post HBV infection of the dHepaRG cells, the cells were

922

mock-treated or treated with 200nM cGAMP for an IRF3 nuclear translocation assay

923

by immunofluorescence (left part). The number of cells with or without nuclear IRF-3

924

from HBs (-) and HBs (+) hepatocyte-like cells in three different visual fields at low

925

magnification was counted and presented as the mean percentage + SD (right part). (I)

926

PHHs were seeded onto the chambered coverglass or 24-well plates. At day 10 post

927

HBV infection, the cells were either collected for the immunofluorescence (left part)

928

or mock-treated or treated with 200nM of cGAMP for 6h and then harvested for the

929

measurement of IFN-β mRNA by quantitative RT-PCR (right part). The results are

930

representative of three independent experiments.* P

Hepatitis B virus polymerase disrupts K63-linked ubiquitination of STING to block innate cytosolic DNA-sensing pathways.

The cellular innate immune system recognizing pathogen infection is essential for host defense against viruses. In parallel, viruses have developed a ...
6MB Sizes 2 Downloads 4 Views