Downloaded - American Society for Microbiology

2 downloads 0 Views 3MB Size Report
Dec 10, 2014 - LDs were stained with HCS LipidTOXTM RED (Invitrogen), and the ...... immunofluorescence with anti-Core and HCS LipidTOX™ Red (LD).
JVI Accepts, published online ahead of print on 10 December 2014 J. Virol. doi:10.1128/JVI.02982-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Phosphatidylserine-Specific Phospholipase A1 is involved in Hepatitis C Virus

2

Assembly through NS2 Complex Formation

3

Min Guoa, b, Rongjuan Peia*, Qi Yanga, b, Huang Caoa, b, Yun Wanga, Chunchen Wua,

4

Jizheng Chena, Yuan Zhoua, Xue Hua, Mengji Luc, Xinwen Chena*

5

a. State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy

6

of Sciences, Wuhan, 430071,China,

7

b. University of Chinese Academy of Sciences, Beijing, China

8

c. Department of Infectious Disease, University hospital Essen, University of

9

Duisburg-Essen, Essen, Germany

10

Running title, role of PLA1A in HCV assembly

11

12

Corresponding author:

13

Prof. Dr. Xinwen Chen

14

State Key Laboratory of Virology,

15

Wuhan Institute of Virology, Chinese Academy of Sciences,

16

Wuhan 430071, People’s Republic of China

17

Phone&Fax: 86-027-87199106;

18

E-mail: [email protected]. cn

19 20

Dr. Rongjuan Pei

21

State Key Laboratory of Virology,

22

Wuhan Institute of Virology, Chinese Academy of Sciences, 1

23

Wuhan 430071, People’s Republic of China

24

Phone: 86-027-87197575;

25

E-mail: [email protected]. cn

26

2

27

ABSTRACT

28

Several members of the phospholipase family have been reported to be involved in

29

hepatitis C virus (HCV) replication. Here, we identified another phospholipase,

30

phosphatidylserine-specific phospholipase A1 (PLA1A), as a host factor involved in

31

HCV assembly. PLA1A was upregulated by HCV infection; and PLA1A knockdown

32

significantly reduced J399EM (genotype 2a) HCV propagation at the assembly but

33

not the entry, RNA replication, and protein translation steps of the life cycle. Protein

34

localization and interaction analysis further reveal a role of PLA1A in the interaction

35

of NS2-E2 and NS2-NS5A, as the formation of the NS2-E2 and NS2-NS5A

36

complexes was weakened in the absence of PLA1A. In addition, PLA1A stabilized

37

the NS2/NS5A-dotted structure during infection. These data suggest that PLA1A

38

plays an important role of bridging the membrane-associated NS2-E2 complex and

39

the NS5A-associated replication complex via its interaction with E2, NS2 and NS5A,

40

which leads to a coordinating interaction between the structural and non-structural

41

proteins and facilitates viral assembly.

42 43

Importance

44

Hepatitis C virus (HCV) genomic replication is driven by the replication complex and

45

occurs at the membranous web, while the lipid droplet is the organelle in which virion

46

assembly is initiated. In this study, we identified phosphatidylserine-specific

47

phospholipase A1 (PLA1A), a member of phospholipase A 1 family, as a novel host

48

factor involved in the assembly process of HCV. PLA1A which is induced by HCV 3

49

infection at late infection stage interacts with HCV E2, NS2 and NS5A proteins and

50

enhance and stabilize the NS2-E2 and NS2-NS5A complex formation, which is

51

essential for viral assembly. Thus, PLA1A is an important host factor which is

52

involved in the initiation of the viral assembly in close proximity to Core-decorated

53

lipid droplet through bringing together the HCV replication complex and envelope

54

complex.

55

4

56

Introduction

57

Hepatitis C virus (HCV) is a major cause of chronic liver disease, affecting

58

approximately 185 million people worldwide (42). HCV is a positive single-stranded

59

RNA virus belonging to the Flaviviridae family. The HCV 9.6 kb genome contains a

60

large open reading frame encoding a single polyprotein that is processed into its

61

structural proteins (Core, E1 and E2) and nonstructural (NS) proteins (p7, NS2, NS3,

62

NS4A, NS4B, NS5A and NS5B) by host and viral proteinases (24). The structural

63

proteins are components of the virion, while the non-structural proteins NS3 to NS5B

64

compose the minimal viral replicase governing RNA replication (4, 25).

65 66

The overall HCV life cycle has been well defined since the development of an

67

infectious HCV cell culture system (23, 49, 51). HCV genomic replication is driven

68

by the replication complex (RC) and occurs at the membranous web, a rearranged

69

membrane structure induced by virus infection (9, 12, 43). Recent progress regarding

70

the study of the assembly process demonstrates that the lipid droplet (LD) is the

71

organelle in which virion assembly is initiated (29) and that, in addition to the

72

structural proteins, almost all the nonstructural proteins and many host factors are

73

involved in this process (22). The roles of NS2 and P7 in virion assembly attract

74

attention because these molecules are not components of the virion or the replicase.

75

NS2 is a polytopic transmembrane protein containing 3 putative transmembrane

76

segments (17) and is suggested to serve as the scaffold for virus assembly by

77

interacting with both structural and nonstructural proteins such as E1/E2, NS3/4A and

78

NS5A (26, 37, 44). P7 is a small protein with 63 amino acids (aa) harboring ion 5

79

channel activity. Its role in HCV morphogenesis has been studied by mutation

80

analysis and has been shown to be important for capsid assembly and envelopment (6,

81

19, 45). NS5A is another critical factor in the assembly process and is recruited to the

82

Core-decorated LD through the interaction between its domain II and Core, bringing

83

HCV RNA to the assembly site (27).

84 85

In addition to viral proteins, several host factors participate in the HCV assembly

86

process by influencing the localization of HCV proteins or by mediating the

87

interactions

88

diacylglycerol acyltransferase-1 (DGAT1) was first found to recruit the Core protein

89

to LD (14). Recently, the interaction between DGAT1 and NS5A was confirmed and

90

was shown to be the bridge between Core and NS5A, facilitating HCV assembly (7,

91

14). Two additional proteins, Rab18 and TIP47, were shown to interact with NS5A

92

and promote the interaction between viral replication sites and LD (36, 38, 48). In

93

addition, signal peptidase complex subunit 1 (SPCS1) facilitates the interaction

94

between NS2 and E2 and is involved in the early step of the assembly of infectious

95

particles (46).

between

HCV

proteins.

The

triglyceride-synthesizing

enzyme

96 97

Several members of the phospholipase A2 family such as PLA2G4A, PLA2G4C and

98

PLA2GXIIB are involved in HCV replication via various mechanisms (21, 28, 39, 50),

99

implying a role for the phospholipase family in HCV replication. Here, we identified

100

another

member

of

the

phospholipase

family,

phosphatidylserine

specific 6

101

phospholipase A1 (PLA1A), as a host factor involved in HCV assembly. PLA1A was

102

first identified in rat pellets (39), and PLA1A mRNA is present at high levels in

103

human liver tissue (2). PLA1A specifically acts on phosphatidylserine (PS) and

104

1-acyl-2-lysophosphatidylserine (lyso-PS) to hydrolyze fatty acids at the sn-1 position

105

of these phospholipids (30). PLA1A is a secreted protein, and its substrate, PS,

106

typically localizes to the inner leaflet of the lipid bilayer, and when PS is exposed on

107

the surface of the cell membrane, PLA1A exerts its lipase functions. Although the

108

lipase activity of PLA1A has been verified, its functions in vivo still remain unknown.

109

However, PLAIA was shown to act with its substrate PS and product lysoPS (30),

110

which is a lipid mediator that plays a role in the inflammation response (10).

111 112

In the present study, we revealed a role for PLA1A in HCV assembly. PLA1A

113

interacts with the HCV E2, NS2 and NS5A proteins, facilitating NS2-E2 and

114

NS2-NS5A complex formation during virus assembly.

115

7

116

Materials and methods

117

Cell culture

118

Huh7.5.1 (kindly provided by Prof. Frank Chisari) and Huh7 cells were cultured in

119

Dulbecco’s modified Eagle medium (DMEM) (Invitrogen) supplemented with 2 mM

120

L-glutamine, nonessential amino acids, and 10% fetal bovine serum (FBS)

121

(Invitrogen). The subgenomic HCV replicon cell lines (Huh7.5.1-SGR (50) and Con1

122

(33) containing the subgenomic genotype 2a and 1b HCV, respectively) were grown

123

in the same medium supplemented with 500 μg ml-1 G418.

124 125

Virus production

126

The JFH-1 virus used in this study was based on the pJFH-1 plasmid kindly provided

127

by Prof. Wakita (49). The J399EM strain was derived from the JFH-1 virus by

128

insertion of eGFP into the HCV NS5A region (13). The JFH1-HA virus with an HA

129

epitope at the N-terminus of NS2 was constructed as previously described (37). The

130

viral titer in the culture supernatants and cell lysates was determined by a modified

131

end-point dilution assay as previously described (33, 50).

132 133

Plasmid construction

134

The PLA1A (NM_001206960) coding sequence was amplified by PCR and inserted

135

into the pXJ40-HA and pXJ40-Flag plasmids. To generate the HA- or Flag-tagged

136

HCV protein expression plasmids, the coding sequences of HCV Core, E1, E2, NS2,

137

NS3, NS34A, NS4B, NS5A and NS5B of genotype 2a (JFH1, AB047639) were 8

138

amplified and inserted into the pXJ40-HA and pXJ40-Flag vectors. The bicistronic

139

reporter, pHCV-IRES, was previously described (50).

140 141

RNA interference

142

The following siRNAs were used: siRNAs specific to PLA1A (siPLA1A-2,

143

D-008411-02-0002; siPLA1A-3, D-008411-03-0003; siPLA1A-4, D-008411-04-0004,

144

Thermo Scientific Dharmacon), AllStars Negative Control siRNA (siControl,

145

#1027281, QIAGEN) and siRNA specific to HCV (siHCV, target sequence: 5 -

146

GGUCUCGUAGACCGUGCAC - 3). The siRNAs were transfected using

147

Lipofectamine 2000 (Invitrogen) at a final concentration of 20 nM according to the

148

manufacturer’s instructions. To maintain the gene silencing effect from the beginning

149

of infection to the last time point analyzed, the cells were split at 24 hours

150

post-transfection (hpt) and transfected with the same siRNA again. HCV infection

151

was performed 6 h after the second transfection.

152 153

Stable cell line construction

154

The coding sequences for short hairpin RNA (shRNA, shPLA1A#4 5’-

155

GGTTTCCTTTGCCGATCTTAT - 3’) targeting the PLA1A gene and a negative

156

control shRNA (5’- GGTGCAGCAGATTGTGAATACCCATAGTAA - 3’) were

157

cloned into the shRNA expression vector pSUPER.retro.neo (OligoEngine, Inc)

158

following the manufacturer’s instructions. The retroviruses were produced in 293T

159

cells by co-transfection of the shPLA1A or shNC plasmid, pVPack-GP and 9

160

pVPack-VSV-G (Stratagene). The retrovirus-containing supernatants were harvested

161

at 72 hpt. To generate the PLA1A knockdown cell line, the Huh7.5.1 cells were

162

transduced with the retrovirus in the presence of 8 μg ml-1 polybrene (Sigma), and

163

stable knockdown pools were isolated by G418 selection and named Huh7.5.1-shNC

164

(or shNC) and Huh7.5.1-shPLA1A#4 (or shPLA1A#4).

165 166

Western blot, co-immunoprecipitation analysis and indirect immunofluorescence

167

staining

168

Whole cell lysates were prepared and quantified using the Bradford method (BioRad

169

#500-0006). Equal amounts of protein samples (30 μg) were subjected to SDS-PAGE

170

and transferred onto a nitrocellulose filter membrane (Millipore). After blocking with

171

5% non-fat milk in TBST, the membranes were incubated with specific primary

172

antibodies and corresponding peroxidase-conjugated secondary antibodies. The

173

following antibodies were used for western blot: anti-NS3 (#ab65407, Abcam),

174

anti-Core (#ab2740, Abcam), anti-Flag (#F1084, Sigma), anti-HA (#H9658, Sigma),

175

anti-PLA1A (produced by Abmart), and anti-beta-actin (#sc-47778, Santa Cruz). The

176

proteins were visualized using suitable HRP-conjugated secondary antibodies

177

(Jackson Immuno Research) and SuperSignal-Femto chemiluminescent substrate

178

(Pierce).

179 180

Co-immunoprecipitation and indirect immunofluorescence staining were performed

181

as previously described (50). The Alexa Flour 561/488/633-conjugated secondary 10

182

antibodies used for indirect immunofluorescence staining were obtained from

183

Invitrogen. LDs were stained with HCS LipidTOXTM RED (Invitrogen), and the

184

nuclei were stained using Hoechst33258 (Invitrogen).

185 186

Quantitative real-time RT-PCR

187

Total RNA from cultured cells was extracted using TRIzol reagent (Invitrogen) and

188

digested with RNase-free DNase (Promega) according to the manufacturer’s protocols.

189

HCV RNA in the supernatant was prepared using TRIzol LS reagent (Invitrogen).

190

Specific mRNAs and HCV RNAs were quantified by one-step real-time RT-PCR

191

using the QuantiFast SYBR Green RT-PCR kit (Qiagen). The mRNA and HCV RNA

192

levels were normalized against the copy number of human beta-actin mRNAs. The

193

forward

194

GGAACTGAGAAACAAGGACACC

195

AAACTCGGTTGGAAGACTGAAA - 3’, and the primers for HCV and actin

196

were previously described (52).

and

reverse

primers

used

to -

amplify

PLA1A 3’

and

were 5’

5’

- -

197 198

Membrane flotation fractionation

199

For cell fractionation, 9 ×107 cells were resuspended in 1 ml of cold hypotonic buffer

200

(10 mM Tris-HCl, pH 7.8, 10 mM NaCl, EDTA-free protease inhibitor cocktail

201

(Roche)) and mechanically disintegrated by forcing the cell suspension through a

202

25-gauge injection needle 30 times. The homogenate was centrifuged at 900 × g for 5

203

min to pellet the nuclear fraction, followed by centrifugation at 15,000 × g for 20 min 11

204

to pellet the cell membrane fraction. The deposit contained the cytoplasmic fraction,

205

and the cell membrane fraction was resuspended in 950 μl hypotonic buffer, NP40

206

was added to a final concentration of 1%, and the solution was incubated for 30 min

207

on ice.

208 209

Regarding sucrose density gradient centrifugation, 1 ml aliquots treated as above were

210

mixed with 1 ml 72% (wt/v) sucrose in Laemmli sample buffer (LSB; 50 mM

211

Tris-HCl, pH 7.5, 25 mM KCl, 5 mM MgCl2) in an ultracentrifuge tube and overlaid

212

with 4 ml 55% sucrose and 1.5 ml of 10% sucrose. The aliquots were then subjected

213

to ultracentrifugation (38,000 × g for 16 h), and every 1 ml fraction was collected

214

from the top of the gradient, mixed with 4 ml cold methyl alcohol and centrifuged at

215

10,000 × g for 10 min to pellet the membrane proteins. The protein samples were

216

characterized using SDS-PAGE and immunoblotting.

217 218

Proteolytic digestion protection assay

219

HCV-infected cells seeded in 6-well dishes were collected in 170 μl proteinase K

220

buffer (50 mM Tris-HCl, pH 8.0, 10 mM CaCl2, 1 mM DTT) and subjected to five

221

freeze-thaw cycles. Next, 50 μl of the crude lysate was left untreated, 50 μl was

222

treated with 50 μg ml-1 proteinase K (Qiagen) for 20 min on ice, and another 50 μl

223

was lysed with 5% (v/v) Triton X-100 prior to proteinase K treatment. Proteinase K

224

digestion was terminated by the addition of 5 mM PMSF on ice for 10 min.

225

Subsequently, 13 μl of 5  SDS sample buffer was added, and the sample was heated 12

226

to 95°C for 10 min. The amount of residual core protein was determined by

227

SDS-PAGE and immunoblotting.

228 229

Statistical analysis

230

Data were analyzed using a two-tailed unpaired t-test. P-values were calculated, and

231

statistical significance was reported as highly significant with *(p