JVI Accepts, published online ahead of print on 26 November 2014 J. Virol. doi:10.1128/JVI.03106-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Hepatitis B core protein sensitizes hepatocytes to tumor necrosis factor-induced apoptosis

2

by suppression of the phosphorylation of mitogen-activated protein kinase kinase 7

3

Baosen Jia,a Minggao Guo,b Gaiyun Li,a Demin Yu,c Xinxin Zhang,c Ke Lan,a# Qiang Denga#

4

Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese

5

Academy of Sciences, Shanghai, China a

6

Department of General Surgery, Sixth People’s Hospital, Shanghai Jiaotong University School

7

of Medicine, Shanghai, China b

8

Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of

9

Medicine, Shanghai, China d

10

#Address correspondence to Dr. Qiang Deng, E-mail: [email protected]; or Dr. Ke Lan,

11

E-Mail, [email protected]

12

Running Title:

13

HBc is pro-apoptotic by blocking MKK7

14

1

15

Abstract

16

Hepatitis B, which caused by Hepatitis B virus (HBV) infection, remains a major health threat

17

worldwide. Hepatic injury and regeneration from chronic inflammation is the main driving

18

factor of liver fibrosis and cirrhosis in chronic hepatitis B. Pro-inflammatory tumor necrosis

19

factor (TNF)-α has been implicated as a major inducer of liver cell death during viral hepatitis.

20

Here, we report that in hepatoma cell lines and in primary mouse and human hepatocytes,

21

expression of hepatitis B core (HBc) protein made cells susceptible to TNF-α-induced

22

apoptosis. We found that receptor of activated protein kinase C (RACK) 1 interacted with HBc

23

by tandem affinity purification and mass spectrometry. RACK1 was recently reported as a

24

scaffold protein that facilitates the phosphorylation of mitogen-activated protein kinase kinase

25

7 (MKK7) by its upstream activators. Our study showed that HBc abrogated the interaction

26

between MKK7 and RACK1 by competitively binding to RACK1, thereby downregulating

27

TNF-α-induced phosphorylation of MKK7 and the activation of c-Jun N-terminal kinase. In

28

line with this finding, specific knockdown of MKK7 increased the sensitivity of hepatocytes to

29

TNF-α-induced apoptosis, while overexpression of RACK1 counteracted the pro-apoptotic

30

activity of HBc. Capsid particle formation was not obligatory for HBc pro-apoptotic activity, as

31

analyzed using an assembly-defective HBc mutant. In conclusion, the expression of HBc

32

sensitized hepatocytes to TNF-α-induced apoptosis by disrupting the interaction between

33

MKK7 and RACK1. Our study is thus a first indication of the pathogenic effects of HBc in

34

liver injury during hepatitis B.

35

Importance:

36

Our study revealed a previously unappreciated role of HBc in TNF-α-mediated apoptosis. The

37

pro-apoptotic activity of HBc is important for understanding hepatitis B pathogenesis. In

38

particular, severe hepatitis-associated HBV variants may upregulate apoptosis of hepatocytes 2

39

through enhanced HBc expression. Our study also found that MKK7 is centrally involved in

40

TNF-α-induced hepatocyte apoptosis and revealed a multifaceted role for JNK signaling in this

41

process.

42

Key words:

43

Hepatitis B core protein; apoptosis; TNF-α; MKK7; RACK1

44

3

45

Introduction

46

Hepatitis B virus (HBV) infection remains a major public health threat affecting an estimated

47

400 million individuals worldwide, with high risk of developing severe liver diseases,

48

including cirrhosis and hepatocellular carcinoma. The host protective immune response is also

49

responsible for liver pathogenesis during HBV infection.

50

Pro-inflammatory tumor necrosis factor (TNF)-α is critical for controlling HBV in clinical

51

settings and in model systems, possibly by destabilizing cytoplasmic viral nucleocapids and

52

reducing nuclear viral DNA (1, 2). Recently, TNF-α was reported to reduce host cell

53

susceptibility to HBV entry via activation-induced cytidine deaminase (3). On the other hand,

54

TNF-α has also been implicated as a mechanism of hepatic injury through induction of cellular

55

apoptosis during viral hepatitis (4-9). Apoptosis is organized, genetically controlled

56

programmed cell death. Clearance of apoptotic debris stimulates transforming growth factor-β

57

expression and induces collagen I secretion by hepatic stellate cells. These processes have been

58

suggested as the core machinery for liver fibrogenesis and cirrhosis (10, 11).

59

Overexpressing HBV core (HBc), precore, X, small and middle envelope proteins (S2S), as

60

well as the viral replication, is not associated with liver disease in transgenic mouse models

61

(12). Hence HBV itself is not considered to be directly cytopathic for infected hepatocytes.

62

This notion has to be re-examined, however, in a pathophysiological setting. Of particular

63

interests, naturally occurring mutations within the basic core promoter of HBV are implicated

64

in severe liver diseases including fulminant hepatitis, which results in up to 15-fold increased

65

HBc synthesis (13-17). The fulminant hepatitis-associated mutant induces apoptosis in primary

66

hepatocytes and is not reversed by inhibition with nucleoside analogs (15). This finding

67

suggests that HBc might be directly responsible for virus-induced hepatocyte death, but the

68

underlying mechanism remains undefined.

4

69

HBc is a 21 kDa viral structural protein with an N-terminal domain that is responsible for viral

70

capsid assembly and a C-terminal arginine-rich polypeptide that interacts with HBV

71

pregenomic RNA. HBc is found to be distributed in both the nucleus and the cytoplasm in

72

HBV-producing hepatocytes and transgenic mice. Clinically, a nucleus-dominant distribution

73

of HBc is associated with minor hepatitis activity while a cytoplasmic distribution of HBc is

74

associated with chronic active liver disease (18). In the present study, we report that expression

75

of HBc increased susceptibility of hepatocytes to TNF-α-induced apoptosis by disrupting the

76

interaction between receptor for activated c kinase 1 (RACK1) and mitogen-activated protein

77

kinase kinase 7 (MKK7) (19), leading to downregulation of TNF-α-induced phosphorylation of

78

MKK7 and c-Jun-N-terminal kinase (JNK) (Fig. 1). Our results therefore revealed a previously

79

unappreciated role for HBc in liver pathogenesis during viral hepatitis.

80

5

Materials and Methods

81 82

Plasmid DNA and viral vectors

83

Plasmids encoding HBV Polymerase, X protein, S2S, and HBc, as well as HA-tagged RACK1

84

and Flag-tagged MKK7, were constructed by inserting the appropriate genes into the multiple

85

cloning site (MCS) of pcDNA3.1(-) (Invitrogen). Genes encoding His-tagged RACK1 or

86

His-tagged HBc were subcloned into the MCS of pET-30a(+) (Novagen) for prokaryotic

87

protein expression. For GST pull-down assay, plasmids encoding GST-tagged HBc

88

truncations and GST-tagged RACK1 truncations were constructed by inserting the appropriate

89

genes into the MCS of pGEX-4T-1 (Promega).

90

Recombinant adenovirus (rAd) expressing HBc (rAd-HBc/GFP, with a GFP gene separated by

91

an IRES sequence) was constructed by Obio Technology (Shanghai), using E1-deleted human

92

rAd vector of serotype 5. The lentiviral vector expressing HA-tagged RACK1 was constructed

93

by inserting the sequence into the MCS of pCDH-CMV-MCS-EF1-Puro (SBI) and packaging

94

the expression constructs into pseudoviral particles.

95

siRNAs

96

siRNA oligos against human and mouse MKK7 were synthesized by GenePharma: siMKK7-1:

97

5’-CAGGACAGUUUCCCUACAATT-3’, siMKK7-2: 5’-GCAAGAUGACAGUGGCGAUTT

98

-3’. siRNAs were transfected into cells by Lipofectamine 2000 (Invitrogen). Subsequent

99

treatments were performed 48 hours after transfection.

100

Cells

101

Huh-7, HepG2, HepG2.2.15 and 293T cells were cultured in DMEM containing 10% fetal

102

bovine serum (FBS) and 1% Penicillin-Streptomycin. Primary mouse hepatocytes (PMH) were

103

isolated from male C57BL/6 mice as described below. Primary human hepatocytes (PHH) were

104

from Mucyte Biotech (Nanjing). PMH and PHH were cultured in Williams’ Media E

6

105

containing 10% FBS, 2 mM L-glutamine, 1× nonessential amino acids, and 1%

106

Penicillin-Streptomycin. HepG2 or Huh-7 cells constitutively expressing HBc were constructed

107

by transfecting cells with pcDNA3.1-HBc and selecting with 50 μg/mL Geneticin.

108

Stimulation with TNF-α

109

To induce massive apoptosis, Huh-7 or HepG2 cells were plated into 12-well plates at 0.25-0.5

110

million cells per well. The next day, cells were pretreated with 0.5 μg/mL actinomycin D

111

(Sigma) for 30 minutes and cultured for 14 hours with 100 ng/mL hTNF-α (PeproTech) in the

112

presence of 12 μM (for Huh-7 cells) or 20 μM (for HepG2 cells) of Bay11-7082 (Sigma). For

113

co-treatment with pyrrolidine dithiocarbamic acid (PDTC, Beyotime), 50 μM of PDTC and 10

114

ng/mL hTNF-α were used (for HepG2 cells).

115

The stimulation with TNF-α in PMH and PHH was described previously (27, 28). In brief,

116

cells were pretreated with 0.5 μg/mL actinomycin D for 30 minutes, and cultured with 2 ng/mL

117

of mTNF-α (PeproTech) (for PMH) or 100 ng/ml hTNF-α (for PHH) in the presence of 0.5

118

μg/mL actinomycin D for 14 hours.

119

Isolation of PMH

120

PMH was isolated from C57BL/6 male mice by two step collagenase perfusion technique (29).

121

The study was approved by the Animal Ethics Committee of Institut Pasteur of Shanghai (no.

122

A2012008-2). Living hepatic parenchymal cells were separated by 35% percoll. After seeding

123

for 4-6 hours, cells were refreshed with medium and cultured overnight. The next day, cells

124

were transduced with rAd-HBc/GFP or a control rAd vector (rAd-GFP) at a MOI of 20. After

125

culture for two days, cells were treated with mTNF-α as described above.

126

Apoptosis analysis

127

Apoptosis was assessed 14 hours after stimulation with TNF-α, using PE Annexin V Apoptosis

128

Detection Kit I (BD, 559763) and PE Active Caspase-3 Apoptosis Kit (BD, 550914)

129

respectively according to the manufacturer’s protocol. 7

130

Immunoblot

131

Cell lysates were prepared in radio immunoprecipitation assay (RIPA) buffer (50 mM Tris-HCl,

132

pH 7.4, 150 mM NaCl, 0.5% Triton X-100) containing 1×Protease Inhibitor Cocktail (Roche)

133

and 1×PhosSTOP Phosphatase Inhibitor Cocktail (Roche). Proteins were separated by

134

SDS-PAGE and transferred to PVDF membranes for immunoblotting. Antibodies were against

135

phospho-JNK (Thr183/Tyr185), JNK and MKK7 from Cell Signaling Technology (9251, 9252

136

and 4172). Anti-phospho-MKK7 (Thr275) was from Sigma (SAB4504624). Anti-RACK1 was

137

from BD (610178). Anti-HBc was from Dako (B0586). Antibodies against cleaved caspase 3

138

and poly ADP-ribose polymerase (PARP) were from Cell Signaling Technology (9661 and

139

9542).

140

Tandem affinity purification (TAP)

141

HepG2 cells stably expressing HBc fused with C-terminal Strep and Flag tags were lysed in

142

RIPA buffer containing 1× Protease Inhibitor Cocktail. TAP of Strep and Flag-tagged HBc was

143

performed by two rounds of affinity purification as previously described (30). Elutes were

144

analyzed by SDS-PAGE and mass spectrometry (Shanghai Applied Protein Technology).

145

Co-immunoprecipitation (Co-IP) and GST Pull-down

146

Cells were lysed in RIPA buffer containing 1× Protease Inhibitor Cocktail for 1 hour on ice

147

with brief vortexing every 10 minutes. Lysates were incubated with 1 μg antibody at 4 ºC

148

overnight with gentle rotation. After centrifugation, supernatants were incubated with protein

149

A/G beads (Life Technologies) at 4 ºC for 1 hour. The immunoprecipitates were separated by

150

SDS-PAGE for immunoblotting.

151

GST-fusion proteins or His-tagged proteins were expressed in Escherichia coli BL21, and

152

purified using glutathione-sepharose 4B (GE Healthcare) or nickel-affinity resins (Qiagen)

153

according to the manufacturers’ instructions. For the pull-down assays, GST-fusion proteins

154

were immobilized on glutathione beads and incubated with purified His-tagged proteins in 8

155

RIPA buffer containing 0.5% BSA, at 4 ºC overnight with gentle rotation. Bound proteins were

156

analyzed by SDS–PAGE and immunoblotting.

157

Immunofluorescence assay

158

Cells were fixed with 4% paraformaldehyde in Phosphate buffered saline (PBS) at room

159

temperature for 15 minutes followed by permeabilization with 0.1% Triton X-100. After

160

blocking with 5% goat serum, cells were incubated with 1:250 anti-RACK1 (BD, 610178) and

161

1:200 anti-HBc (Dako, B0586) in PBS containing 1% goat serum and 0.05% Triton X-100 at 4

162

ºC overnight. Alexa Fluor 555 Goat Anti-Mouse antibody and Alexa Fluor 488 Goat

163

Anti-Rabbit antibody (Life Technologies) were used as secondary antibodies for anti-HBc or

164

anti-RACK1 staining respectively.

165

Statistics

166

Data are expressed as means ± standard error of the mean (SEM). Student’s unpaired t-tests

167

were performed with GraphPad Prism software.

168

9

Results

169 170

HBc increases TNF-α-induced apoptosis in hepatoma cells and primary hepatocytes

171

TNF-α is implicated in hepatic injury from a variety of liver diseases. In the presence of the

172

NF-κB inhibitor Bay11-7082 (Fig. S1), stimulation with TNF-α significantly increased the

173

percentage of annexin V-binding HepG2.2.15 cells compared with HBV-free HepG2 cells.

174

Treatment with lamivudine did not significantly alter annexin V-binding to HepG2.2.15 cells,

175

suggesting that the viral replication had little effect on TNF-induced apoptosis (Fig. 2 A-B).

176

Upon TNF-α/Bay11-7082 treatment, annexin V-binding increased in Huh-7 cells that were

177

constitutively expressing HBc, but not in cells with expression of the other viral proteins (Fig.

178

2C). Expression of all these viral proteins could be readily detected by specific antibodies

179

respectively (data not shown), although they were likely to be at different levels. Mutant assays

180

on the HBV genome also revealed that HBc might be directly responsible for increased

181

apoptosis in HBV-producing cells (Fig. 2D). In either HepG2 (Fig. 2E) or Huh-7 cells (data

182

not shown), ectopic expression of HBc increased the production of active caspase 3, a typical

183

marker of apoptosis.

184

rAd vectors were constructed for transduction assays in primary mouse or human hepatocytes.

185

In the presence of actinomycin D (27, 28), an inhibitor of de novo gene transcription, TNF-α

186

treatment upregulated active caspase 3 and increased apoptosis in the hepatocytes transduced

187

with rAd-HBc/GFP, compared with that in rAd-GFP transduced cells (Fig. 2F-G). Collectively,

188

these data indicated that HBc promoted TNF-α-induced apoptosis in cultured hepatoma cells

189

and primary hepatocytes.

190

HBc downregulates TNF-α-induced MKK7 and JNK phosphorylation

191

TNF-α is a conditional death ligand. Its apoptotic capability is largely modulated by the NF-κB

192

and JNK signaling cascades (Fig. 1). HBc did not affect NF-κB signaling pathway activity as

10

193

measured by reporter assays (Fig. 3A). However, TNF-α-induced phosphorylation of MKK7

194

and downstream JNK activation were substantially suppressed in HepG2 (Fig. 3B) and Huh-7

195

(Fig. 3C) cells that constitutively expressed HBc, compared to HBc-negative cells. TNF-α also

196

induced phosphorylation of MKK4 in Huh-7 cells, but this was not affected by HBc (Fig. 3C).

197

The inhibitory effect of HBc on TNF-α-induced MKK7/JNK activation occurred also in the

198

context of an intact HBV genome. TNF-α-stimulated MKK7/JNK activation was reduced in

199

HepG2 cells expressing the 1.2mer overlength HBV genome, but not in cells with an HBc-null

200

mutant genome (Fig. 3D). These data clearly indicated that expression of HBc blocked

201

TNF-α-stimulated activation of MKK7/JNK.

202

Pro-apoptotic activity of HBc is mediated by blockage of MKK7 phosphorylation

203

TNF-α alone stimulated rapid, transient activation of JNK (within 1 hour, Fig. 3), and does not

204

cause apoptosis (data not shown). In the presence of the NF-κB inhibitor, however, TNF-α

205

stimulation induced prolonged phosphorylation of MKK7/JNK (Fig. 4A). This prolonged JNK

206

activation is thought to be essential in regulating TNF-induced apoptosis, although its precise

207

role in this process remains controversial (20, 22, 24-26). While the activation of MKK7/JNK

208

decreased from hours 8 to12, cleaved caspase 3 and PARP were detected as early as 4 hours

209

after stimulation, indicating the onset of programmed cell death (Fig. 4A). Of particular

210

interests, the prolonged MKK7/JNK activation was remarkably suppressed (mainly limited to

211

1-4 hours after stimulation) in HBc-expressing HepG2 cells that underwent significantly

212

enhanced apoptosis (Fig. 4A, B).

213

We then tested whether inactivation of MKK7/JNK was directly responsible for

214

TNF-α-induced apoptosis. This was achieved by knockdown of MKK7 expression through

215

RNA interference. HepG2 cells transfected with MKK7-specific siRNAs had increased

216

apoptosis upon stimulation with TNF-α (Fig. 4C). Similar results were obtained by knockdown

217

of MKK7 expression in mouse primary hepatocytes (Fig. 4D). Collectively, these data

11

218

suggested that the pro-apoptotic activity of HBc was achieved by blocking MKK7 activation.

219

RACK1 is a cellular protein that interacts with HBc

220

To understand the mechanism of the pro-apoptotic activity of HBc, we performed tandem

221

affinity purification to identify potential cellular interacting protein(s). RACK1, a multifaceted

222

scaffold protein, was identified as a candidate for HBc interaction by mass spectrometry. In

223

293T cells, HA-tagged RACK1 co-immunoprecipitated with Flag-tagged HBc, and vise versa

224

(Fig. 5A, B). In HepG2.2.15 cells that stably produce HBV, endogenous RACK1

225

co-immunoprecipitated with HBc (Fig. 5C). Consistent with their physical interaction, both

226

HBc and endogenous RACK1 were distributed throughout the cytoplasm of HepG2.2.15 cells

227

(Fig. 5D).

228

HBc has an N-terminal domain (aa1–149) and a C-terminal arginine-rich polypeptide

229

(aa150–183) (31), while RACK1 contains 7 Trp-Asp (WD) repeats (32). Interaction between

230

HBc and RACK1 was further verified by GST pull-down assay. In particular, His-tagged

231

RACK1 co-precipitated with the GST-fused HBc N-terminal domain, but not the HBc

232

C-terminal polypeptide (Fig. 5E), while His-tagged HBc co-precipitated only with GST-fused

233

RACK1 WD6-7, but not WD1-3 or WD1-5 (Fig. 5F, G). Neither GST-fused RACK1 WD6 nor

234

WD7 pulled down His-tagged HBc (Fig. 5G). These data indicated that RACK1 bound the

235

HBc N-terminal domain directly through the WD6-7 region, probably through the RACK1

236

knob structure between WD6 and WD7 domains (32).

237

HBc pro-apoptotic activity is independent of capsid particle formation

238

HBc is a structural protein of HBV and assembles into icosahedral capsid particles. We thus

239

investigated if free monomeric or dimeric protein, or the capsid particle accounted for HBc

240

pro-apoptotic activity.

241

We used an HBc mutant with a tyrosine-to-alanine substitution at aa 132 (Y132A). This

242

mutation prevents HBc from self-assembly into capsid particles but retains ability to form 12

243

homodimers (33, 34). As revealed by native gel electrophoresis assays, ectopically expressed

244

wild-type HBc formed capsid particles that were absent in cells transfected with the HBc

245

mutant (Fig. 6A). In contrast to previous reports (1, 2), TNF-α treatment, with or without

246

NF-κB inhibitor, did not significantly reduce capsid particles in the cytosol. This result

247

probably reflected a difference between the genuine nucleocapsids and the empty capsids that

248

lack HBV DNA/RNA (35). Expression of HBc Y132A was detected by SDS-PAGE with

249

immunoblotting, albeit with a relatively lower level compared to that of wild-type HBc. In the

250

presence of TNF-α/Bay11-7082, expression of the assembly-defective HBc mutant displayed

251

similar cytotoxicity to wild-type HBc (Fig. 6B). In this regard, the HBc mutant engaged in a

252

direct interaction with RACK1 (Fig. 6C). These data clearly suggested that the pro-apoptotic

253

activity of HBc was independent of capsid formation.

254

HBc and MKK7 bind RACK1 competitively

255

RACK1 is reported to interact with MKK7 through its WD6-7 region, which facilitates MKK7

256

phosphorylation by upstream MAP3Ks (19). This suggested that HBc and MKK7 might bind

257

RACK1 competitively.

258

RACK1/MKK7 interaction was confirmed in our study by a GST pull-down assay. His-tagged

259

RACK1 co-precipitated with the GST-fused MKK7, which however was largely blocked in a

260

dose-dependent manner by co-incubation with purified His-tagged HBc (Fig. 7A). In 293T

261

cells expressing Flag-tagged-MKK7 (MKK7-Flag), co-expression of HBc led to a significant

262

decrease in the amount of MKK7-Flag co-precipitated RACK1 (about 4-fold, Fig. 7B).

263

Moreover, in HepG2 cells with high endogeneous expression of MKK7, RACK1 and

264

transforming growth factor beta-activated kinase 1 (TAK1, an MAP3K that phosphorylates

265

MKK7 upon TNF-α stimulation) co-immunoprecipitated with MKK7. Those interactions were

266

barely detectable when HBc was expressed (Fig. 7C). The proteins RACK1, TAK1, MKK7

267

and JNK were all detected in PHH, but with lower levels than in HepG2 or Huh-7 cells (Fig.

13

268

S4). Our results therefore indicated that HBc disrupted interaction between MKK7 and

269

RACK1, which might abrogate TNF-α-mediated MKK7/JNK activation.

270

Pro-apoptotic activity of HBc is reversed by RACK1 overexpression

271

Competitively binding to RACK1 thus appeared to be the means by which HBc affected

272

TNF-α-induced apoptosis. This hypothesis was verified by overexpression of RACK1 to

273

counteract the effect of HBc. As shown in Fig. 8A, expression of HBc in HepG2 cells resulted

274

in reduced MKK7/JNK phosphorylation upon TNF-α stimulation, which however was reversed

275

by ectopic expression of HA-tagged RACK1 in these cells through lentiviral transduction.

276

Moreover, in the presence of NF-κB inhibitor Bay11-7082 (data not shown) or PDTC (Fig. 8B),

277

overexpression of RACK1 prevented the upregulation of TNF-α-induced apoptosis by HBc in

278

these cells. These data suggested that HBc suppressed TNF-α-mediated MKK7/JNK activation

279

and promoted apoptosis mainly through RACK1 blocking.

280

14

281

Discussion

282

TNF-α mediates both cell survival and cell death signaling through TNF receptor-1, by

283

activation of NF-κB and caspase-8 respectively (Fig. 1). Alteration in NF-κB signaling largely

284

accounts for the change in TNF-α function from anti-apoptotic to pro-apoptotic. However, the

285

underlying mechanism and the physiological relevance of the NF-κB signaling alteration are

286

poorly understood. Nevertheless, TNF-mediated apoptosis can be experimentally induced in

287

the setting of global transcriptional or translational arrest, or selective inactivation of NF-κB

288

(21, 22, 24). Our study took advantage of these experimental conditions, based on previous

289

studies by others, to investigate the potential role of HBc in TNF-α-induced apoptosis.

290

Bay11-7082 has been widely used as a tool compound that inhibits NF-κB activation. In our

291

study, NF-κB inhibitor PDTC was also used in order to avoid possible off-target effects of

292

Bay11-7082 (Fig. S2). However, both Bay11-7082 and PDTC are thought to be

293

multi-target-directed compounds, and the mechanisms whereby these compounds affect NF-κB

294

signal transduction are not well-understood.

295

The role of TNF-α must be carefully examined in different pathophysiological settings. For

296

example, TNF-α is implicated in HBV clearance via non-cytotoxic mechanisms in which

297

TNF-α-stimulated NF-κB activation is indispensable (1-3). On the other hand, we propose a

298

scenario for the cytotoxic function of TNF-α (8, 9) in which the NF-κB activation might be

299

coordinately inhibited. For example, multiple apoptotic stimuli including from TNF-α and

300

professional death ligands such as FasL and Apo2L/TRAIL, might function cooperatively

301

during severe hepatitis. The professional death ligands directly induce caspase 8 activation but

302

poorly activate NF-κB (21, 36). It is noted that the apical caspase activation is weak to induce

303

apoptotic death of “type II cells” such as hepatocytes, and requires amplification through

304

crosstalk to the TNF-triggered JNK activation pathway (21, 22, 24). In particular, active

15

305

caspase 8 suppresses TNF-triggered NF-κB signaling, thereby accelerating the programmed

306

cell death (21). We propose that this scenario might occur during severe or fulminant hepatitis,

307

in which HBV mutant-related HBc overexpression is likely to be important in enhancing

308

TNF-α-mediated apoptosis.

309

JNK is an important regulator of TNF-α signaling and a member of the MAP kinase subfamily.

310

Inhibition of de novo protein synthesis or NF-κB activation sensitizes hepatocytes to

311

TNF-α-induced hepatocyte death as a result of sustained JNK activation (22, 24). Initial JNK

312

activation is thought to be transient and associated with cell survival and proliferation through

313

AP-1. Sustained JNK activation and ROS accumulation are associated with apoptotic and

314

necrotic cell death (24). Interestingly, we found that the pro-apoptotic activity of HBc was

315

associated with significant suppression of JNK phosphorylation. Possible explanations could

316

be: i). the precise role of JNK in TNF-α-mediated hepatocyte apoptosis is controversial (24,

317

25). In a recent report, JNK-1 transduced an antiapoptotic signal in TNF-α-mediated

318

hepatocyte apoptosis through Mcl-1 stabilization (26). ii). HBc might not have completely

319

abolished the sustained activation of JNK. MKK7 and MKK4 are both upstream activators of

320

JNK. Phosphorylation of MKK7 but not MKK4 was suppressed during HBc co-expression.

321

However, MKK4 is reported to be only slightly activated by proinflammatory cytokines (37).

322

iii). another possibility is that inactivation of MKK7 itself might be directly pro-apoptotic by

323

an unknown mechanism independent of JNK signal transduction.

324

Direct evidence about MKK7 functions in TNF-α-induced hepatocyte death might come from

325

analysis of a mouse model with a specific genetic abrogation. However, mice lacking MKK7

326

or MKK4 die before birth (38). TAK1 is a direct activator of MKK7 that is critical for the

327

survival of hematopoietic cells and hepatocytes (39). Deletion of TAK1 results in bone marrow

328

and liver failure in mice, due to the massive apoptotic death of hematopoietic cells and

329

hepatocytes (39). Cultures of primary hepatocytes deficient in TAK1 exhibit spontaneous cell 16

330

death that is further increased in response to TNF-α (40). Similar to abrogation of MKK7

331

activation by HBc as described in our study, deletion of TAK1 in hepatocytes abolishes JNK

332

and NF-κB activation and confers susceptibility to TNF-α-mediated cell death.

333

Scaffolding of multicomponent regulatory systems is recognized as a major mechanism for

334

controlling signal transduction pathways (41). Specificity of regulation is achieved by

335

organization of MAPK modules, in part, through scaffolding and anchoring proteins. RACK1

336

is a widely expressed, highly conserved scaffolding protein of 7 WD repeat domains. It

337

interacts with a range of proteins and is involved in many cellular processes (32). A previous

338

study demonstrated that RACK1 is an adaptor for protein kinase C-mediated JNK activation

339

through direct binding to JNK1 and JNK2 (42). More recently, Guo and colleagues reported

340

that RACK1 engages directly with MKK7, enhancing the binding of MKK7 to its upstream

341

MAP3Ks and promoting MKK7 phosphorylation (19). The authors therefore proposed that

342

overexpression of RACK1 augments JNK activity, promoting growth of hepatocellular

343

carcinoma. Our study showed that HBc abrogated TNF-α-induced MKK7 phosphorylation by

344

competitively binding to RACK1. In-depth studies are needed to investigate whether

345

expression of HBc affects development of HBV-related hepatocellular carcinoma.

346

The pro-apoptotic activity of HBc might harm HBV infection. In this regard, induction of

347

apoptosis is an effective way to sacrifice virus-infected host cells and restrict pathogen spread

348

(43). Huang et al. recently found that virus-infected cells use the mitochondrial antiviral

349

signaling protein (MAVS)-MKK7-JNK2 signaling pathway to induce apoptosis as an innate

350

immune protection mechanism (44). HBc therefore could be a pathogen-associated molecular

351

pattern that sensitizes hepatocytes to TNF-α induced apoptosis. Supporting this possibility, Lin

352

et al. (33, 45) recently showed that HBc is a major viral factor for HBV clearance in a

353

hydrodynamics-based mouse model. This effect depends on the self-assembly of HBc into

354

capsid particles, which however is not obligatory for the pro-apoptotic activity of HBc 17

355

described in our present study.

356

HBV is believed to establish a stealthy infection without alerting significant host immunity. In

357

a chimpanzee model with acute viral hepatitis, the proportion of apoptotic and mitotic

358

hepatocytes is lower than 0.3% even at the viral clearance phase, although the turnover rate is

359

virtually accelerated (46). Nevertheless, it is the slow rate of cell destruction and long-term

360

persistent viral infection that cumulates fibrogenesis and cirrhosis in the liver. Apoptosis is

361

thought to be the nexus of liver injury and fibrosis (11). The pro-apoptotic activity of HBc

362

described in our study thus could be critical for understanding the pathogenesis of chronic

363

hepatitis B infection.

364

It will be interesting to discover whether HBc-related mutants of HBV have particular clinical

365

manifestations. Naturally occurring mutations in the HBV basic core promoter provide

366

evidence on this issue. A prevalent HBV variant is a double mutation of C-to-T at nucleotide

367

1768 and a T-to-A change at nucleotide 1770, resulting in highly upregulated core promoter

368

activity. This change is associated with a clinical outbreak of fatal fulminant hepatitis. In

369

particular, this HBV strain induces substantial apoptosis in primary tupaia hepatocytes by a

370

mechanism potentially independent of enhanced replication (15), possibly the strongly

371

upregulated expression of pro-apoptotic HBc.

372

In conclusion, our study found that HBc protein sensitizes hepatocytes to TNF-α-induced

373

apoptosis by competing with MKK7 for binding to RACK1, abrogating MKK7

374

phosphorylation. The study is thus a first indication of that HBc is pathogenic in liver injury

375

during hepatitis B.

376

18

377

Acknowledgement

378

This work was supported by grants from the National Science and Technology Major Projects

379

(2012ZX10002007), Natural Science Foundation (81171566), and National Key Basic

380

Research Program of China (2012CB519000). We also gratefully acknowledge the support of

381

SA-SIBS Scholarship Program.

382

19

Figure Legends

383

384

FIG. 1. Schema of TNF-α signaling pathway that induces apoptosis

385

TNF-α is capable of inducing apoptosis when NF-κB signaling is blocked (20-24). Upon

386

binding to TNF-receptor I, a protein Complex I forms rapidly that activates NF-κB and JNK

387

signaling pathways. Complex I undergoes ligand-dissociated internalization with formation of

388

Complex II. Complex II is composed of FADD and procaspases 8, and triggers apoptosis if

389

NF-κB signaling fails to induce expression of anti-apoptotic genes. JNK is an important

390

regulator of TNF-α-mediated apoptosis (20, 22, 24-26), and is activated via the sequential

391

activation of protein kinases including two MAP kinase kinases (MKK4 and MKK7) and

392

multiple MAP kinase kinase kinases (MAP3Ks). MKK7 is an essential component of the JNK

393

signaling pathway activated by TNF-α. RACK1 is a scaffold protein that facilitates

394

phosphorylation of MKK7 by upstream MAP3Ks (19). It is widely reported that sustained JNK

395

activation mediates TNF-α-induced cell death, although in hepatocytes JNK has both

396

pro-apoptotic and anti-apoptotic aspects (22, 24). We propose that HBc (red) disrupts

397

interactions between MKK7 and RACK1, rendering cells susceptible to TNF-α-mediated

398

apoptosis.

399

FIG. 2. HBc confers susceptibility to TNF-α-induced apoptosis in hepatoma cells and

400

primary hepatocytes

401

(A) Left, annexin V staining of HepG2 or HepG2.2.15 cells 14 hours after co-treatment with

402

TNF-α and Bay11-7082. Cells treated with Bay11-7082 alone, or mock treated, were used as

403

controls. To block HBV replication, HepG2.2.15 cells were pretreated with 20 g/mL

404

Lamivudine (LAM) for 48 hours. Right, representative flow cytometry analysis of cells treated

405

with TNF-α/Bay11-7082. Cells were double-stained with 7-amino actinomycin D (7-AAD) and

406

annexin V. Annexin V positive populations were calculated. (B) Upper, Southern blot of HBV 20

407

replication in HepG2, HepG2.2.15 and LAM-treated HepG2.2.15 cells. Replication

408

intermediates of HBV DNA are RC, relaxed circular; DSL, double-stranded linear; SS, single

409

strand; Lower, HBc levels detected by immunoblotting. (C) Annexin V staining of Huh-7 cells

410

stably transfected with a control plasmid or the plasmid encoding HBc, polymerase (Pol), X

411

protein (HBx), or S2S proteins of HBV respectively. Cells were treated with

412

TNF-α/Bay11-7082 as described in A. (D) Annexin V staining of HepG2 cells stably

413

transfected with a 1.2mer genome of HBV (payw1.2), or mutant HBV genome deficient in

414

HBe and HBc (payw1.2-e/C-null). Mock-transfected cells were used as controls. (E) Left, flow

415

cytometry of active caspase 3 expression after TNF-α/Bay11-7082 treatment in HepG2 cells

416

stably transfected with a control plasmid, or the plasmid encoding HBc (pcDNA3.1-HBc).

417

Right, representative flow cytometry analysis of cells treated with TNF-α/Bay11-7082.

418

Co-treatment with TNF-α and the NF-κB inhibitor PDTC was also tested, with similar results

419

(Fig. S2). (F-G) PMHs (F) or PHHs (G) transduced with rAd-HBc/GFP, or rAd-GFP as control,

420

for 48 hours at a MOI of 20:1. Cells were then treated with mouse or human-derived TNF-α

421

and actinomycin D (ActD), with active caspase 3 expression analyzed by flow cytometry. The

422

error bars indicate SEM. *, P

Hepatitis B virus core protein sensitizes hepatocytes to tumor necrosis factor-induced apoptosis by suppression of the phosphorylation of mitogen-activated protein kinase kinase 7.

Hepatitis B, which caused by hepatitis B virus (HBV) infection, remains a major health threat worldwide. Hepatic injury and regeneration from chronic ...
5MB Sizes 0 Downloads 6 Views