JVI Accepts, published online ahead of print on 25 June 2014 J. Virol. doi:10.1128/JVI.00586-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Title

2

Influenza A virus hemagglutinin and neuraminidase mutually accelerate their

3

apical targeting through clustering of lipid rafts.

4 5

Running title

6

Apical targeting of HA and NA via lipid raft

7 8

Takashi Ohkura,a Fumitaka Momose,a Reiko Ichikawa,a Kaoru Takeuchi,b and Yuko

9

Morikawaa

10

a

11

University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan

12

b

13

of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki 305-8575, Japan

Kitasato Institute for Life Sciences and Graduate School for Infection Control, Kitasato Department of Environmental Microbiology, Division of Biomedical Science, Faculty

14 15

Corresponding author:

16

Yuko Morikawa

17

Kitasato Institute for Life Sciences and Graduate School for Infection Control, Kitasato

18

University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan.

19

Phone:

20

[email protected]

+81-3-5791-6129;

Fax:

+81-3-5791-6268;

E-mail:

21

Abstract

22 23

In polarized epithelial cells, influenza A virus hemagglutinin (HA) and

24

neuraminidase (NA) are intrinsically associated with lipid rafts and target the apical

25

plasma membrane for viral assembly and budding. Previous studies have indicated that

26

the transmembrane domain (TMD) and cytoplasmic tail (CT) of HA and NA were

27

required for association with lipid rafts, but the raft dependencies of their apical

28

targeting are controversial. Here, we show that coexpression of HA with NA accelerated

29

their apical targeting through accumulation in lipid rafts. HA was targeted to the apical

30

even when expressed alone but the kinetics was much slower than that of HA in infected

31

cells. Coexpression experiments revealed that apical targeting of HA and NA was

32

accelerated by their coexpression. The apical targeting of HA was also accelerated by

33

coexpression with M1 but not M2. The mutations in the outer leaflet of the TMD and

34

the deletion of the CT in HA and NA that reduced their association with lipid rafts

35

abolished the acceleration of their apical transport, indicating that the lipid raft

36

association is essential for efficient apical trafficking of HA and NA. An in situ

37

proximity ligation assay (PLA) revealed that HA and NA were accumulated and

38

clustered in the cytoplasmic compartments, only when both were associated with lipid

39

rafts. Analysis with mutant viruses containing nonraft HA/NA confirmed these findings.

40

We further analyzed lipid raft markers by in situ PLA and suggest a possible mechanism

41

of the accelerated apical transport of HA and NA via clustering of lipid rafts.

42

43

Importance

44 45

Lipid rafts serve as sites for viral entry, particle assembly, and budding, leading

46

to efficient viral replication. The influenza A virus utilizes lipid rafts for apical plasma

47

membrane targeting and particle budding. The hemagglutinin (HA) and neuraminidase

48

(NA) of influenza virus, key players for particle assembly, contain determinants for

49

apical sorting and lipid raft association. However, it remains to be elucidated how lipid

50

rafts contribute to the apical trafficking and budding. We investigated the relation of

51

lipid raft association of HA and NA to the efficiency of apical trafficking. We show that

52

coexpression of HA and NA induces their accumulation in lipid rafts and accelerates

53

their apical targeting and suggest that the accelerated apical transport likely occurs by

54

clustering of lipid rafts at the TGN. This finding provides the first evidence that two

55

different raft-associated viral proteins induce lipid raft clustering, thereby accelerating

56

apical trafficking of the viral proteins.

57

Introduction

58 59

Influenza virus is an enveloped, negative-stranded, segmented RNA virus

60

belonging to the Orthomixoviridae family. The virion consists of three integral

61

membrane proteins, hemagglutinin (HA), neuraminidase (NA), and ion channel protein

62

M2. A layer of matrix protein M1 is present underneath the lipid envelope and encases

63

viral ribonucleoprotein (vRNP) complexes. The influenza virus buds from the apical

64

plasma membrane (PM), which is divided by tight junctions in polarized epithelial cells

65

(1). It is considered that all viral components are targeted to the apical PM, where

66

particle budding occurs. HA, NA, and M2 are synthesized at the endoplasmic reticulum

67

(ER) and are transported to the apical PM through the trans-Golgi network (TGN). The

68

apical sorting signals were identified in the transmembrane domain (TMD) of both HA

69

and NA (2, 3). Many studies indicate that during the apical trafficking, HA and NA are

70

associated with lipid raft microdomains, which are enriched in cholesterol and

71

sphingolipids (3, 4), whereas M2 is excluded from these domains (5, 6). Several studies

72

also indicate that the TMD and the cytoplasmic tail (CT) of HA and NA are important

73

for their association with lipid rafts (3, 5, 7). It has been shown that, in the case of HA,

74

palmitoylation at three conserved cysteines in the TMD-CT region is required for

75

association with lipid rafts (8). A very recent study suggested that M2 was a key player

76

in influenza virus particle budding, which is endosomal protein sorting complex

77

required for transport (ESCRT)-independent (9).

78

Lipid rafts are thought to function as platforms for selective concentration of

79

raft-associated proteins to promote protein-protein interactions for their functions (10).

80

Lipid rafts have also been shown to play pivotal roles in apical trafficking in polarized

81

cells (11) and in signal transduction pathways, such as Ras signaling (12) and PIP2

82

signaling (13). It has been suggested that for influenza virus HA and NA, the

83

association with lipid rafts constitutes a part of the machinery necessary for apical

84

trafficking in polarized cells (14, 15). Previous studies have indicated that disruption of

85

lipid rafts by treatment with methyl-β-cyclodextrin (MβCD) and lovastatin delays the

86

TGN-to-apical PM trafficking of HA and missorts to the basolateral membrane, whereas

87

vesicular stomatitis virus G protein, a nonraft-associated basolateral marker, remained

88

unaffected (16), suggesting that raft association is required for apical transport of

89

proteins. However, a number of studies have indicated that some mutations in the HA

90

TMD and CT did not impair apical targeting of HA irrespective of whether or not they

91

caused significant reductions in the raft association (5, 7), suggesting that raft

92

association is not essential for apical transport of HA.

93

Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) are not

94

integral membrane proteins but are associated with lipid rafts through the GPI moieties.

95

In this group of proteins, the raft association is not a determinant for apical sorting

96

because both apical and basolateral GPI-APs are associated with lipid rafts (17).

97

Interestingly, only apical, but not basolateral, GPI-APs form oligomer complexes when

98

they are associated with lipid rafts. When oligomerization of GPI-APs was impaired by

99

mutations, the GPI-APs were missorted to the basolateral PM domain (17). A recent

100

model has suggested that the oligomerization of GPI-APs promotes their stabilization in

101

lipid rafts, leading to their incorporation into apical transport vesicles (10).

102

Influenza virus budding and release require the assembly of viral components,

103

which occurs either during their trafficking to the apical PM or at the stage of particle

104

budding (6, 18, 19). During the trafficking, all viral components, HA, NA, M2, M1, and

105

vRNP, either individually or in their complex forms, are targeted to the apical PM and

106

form a higher order of complex (7, 19, 20). Although the apical sorting determinants for

107

individual viral components have been relatively well studied (2, 3), the molecular

108

mechanisms and kinetics involved in their apical targeting have not been elucidated.

109

In this study, we focused on the kinetics of apical targeting of influenza virus

110

envelope proteins in polarized MDCK cells. We found that HA and NA or M1, but not

111

M2, mutually accelerated their apical PM targeting. Using the TMD-CT mutants of HA

112

and NA, we show that the association of HA and NA with lipid rafts is necessary for the

113

acceleration of their apical PM targeting. Our data indicate that HA and NA come into

114

close proximity (clustering) in lipid rafts upon coexpression. Our data also show that

115

clustering of lipid rafts upon coexpression of HA and NA, suggesting a possible

116

mechanism of accelerated apical transport of HA and NA via the clustering of lipid rafts.

117

Materials and methods

118 119

Viruses and plasmids The H141Y and E142Q mutations were introduced into the HA gene of the

120 121

influenza A/Puerto Rico/8/34 (PR8) virus by inverted PCR. This PR8 derivative was

122

referred to as the wild type (wt) strain in the present study. The authentic PR8 strain was

123

used as the parental virus. The wt and mutant PR8 viruses were generated by reverse

124

genetics system with PolI plasmids (pHH21) and protein expression plasmids, as

125

described previously (21). Briefly, 293T cells were transfected with PolI plasmids for

126

synthesis of each viral RNA segment and protein expression plasmids for PB1, PB2, PA,

127

NP, HA and NA. At 6 h post-transfection (hpt), the cell medium was replaced with

128

Opti-MEM I (Gibco) supplemented with 5 μg/ml acetyl trypsin and 0.3% bovine serum

129

albumin (BSA), and the cells were incubated for 2 or 3 days. Recovered viruses were

130

grown in Madin-Darby canine kidney (MDCK) cells stably expressing HA

131

(MDCK-HA) (the kind gift of Dr. Takizawa from the Institute of Microbial Chemistry,

132

Japan).

133

The HA and NA constructs with deletion of the CT (ΔCT-HA and ΔCT-NA) for

134

recovery of recombinant viruses have essentially been described elsewhere (22, 23). For

135

ΔCT-HA, three consecutive stop codons were placed at the end of the TMD-coding

136

sequence and the downstream nucleotide sequence was left intact. For ΔCT-NA, the

137

authentic start codon was mutated and a new start codon was created at the beginning of

138

the TMD-coding sequence. The constructs were cloned into pHH21. The HA and NA

139

constructs with alanine substitutions in the CT (HA557-559, HA560-563, HA564-566,

140

NA2-3, NA4-6, and NA2-6) and TMD (HA533-535, HA550-552, NA7-10, and

141

NA31-35), and those with cysteine-to-serine substitutions at three palmitoylation sites

142

(HA-SSS), were generated by overlapping PCR and were cloned into pHH21. The

143

viruses with similar alanine substitutions have been described previously (3, 5, 8).

144

The open reading frames of the wt-HA, NA, M1, and M2 genes were cloned

145

into the eukaryotic expression plasmid pCAGGS, respectively. The open reading frames

146

of the HA and NA constructs with deletion of the CT and those with the alanine

147

substitutions were similarly cloned into pCAGGS, respectively. The cDNAs encoding

148

CD59, the 75 kDa neurotrophin receptor (p75), and their GFP-tagged versions

149

(GFP-CD59 and p75-GFP) were also cloned into pCAGGS.

150 151

Cell culture, infection, and DNA transfection

152

MDCK and 293T cells were maintained in Dulbecco’s modified Eagle’s

153

medium (Sigma) supplemented with 10% fetal bovine serum. MDCK cells were seeded

154

into 12-well plates (1×106 cells/well) and were polarized by 12 h-incubation. The

155

polarized MDCK cells were used throughout this study. The MDCK cells were infected

156

with the wt virus at a multiplicity of infection (MOI) of 0.1 or 1 for 1 h. Transfection of

157

DNA was carried out using Lipofectamine LTX (Invitrogen). To synchronize protein

158

expression from plasmids, DNA-Lipofectamine complexes were sedimented by plate

159

centrifugation at 250 × g for 5 min. In some experiments, MDCK cells were transfected

160

with DNA and subsequently superinfected with the authentic PR8 virus.

161 162

Virus growth and plaque assay

163

MDCK cells were inoculated with virus at an MOI of 0.1 in Opti-MEM I

164

supplemented with 0.3% BSA for 1 h at 37°C. After being washed, the cells were

165

incubated in Opti-MEM I supplemented with 5 μg/ml acetyl trypsin and 0.3% BSA. The

166

culture medium was harvested at various time points and was subjected to plaque assay

167

on MDCK or MDCK-HA cells.

168 169

Cholesterol depletion

170

For the inhibition of cholesterol synthesis, 293T and MDCK cells were

171

pre-treated with 8 μM lovastatin (Merck) for 12 h. After transfection, the cells were

172

incubated in the presence of 8 μM lovastatin for 12 or 24 h. The cells were further

173

treated with 5 or 10 mM methyl-β-cyclodextrin (MβCD) (Sigma) in the presence of 8

174

μM lovastatin for 1 h.

175 176

Indirect immunofluorescence assay

177

Polarized MDCK cells were grown on coverslips in 12-well plates and were

178

either infected with virus or transfected with protein expression plasmids. The cells

179

were fixed with 4% paraformaldehyde (PFA) and permeabilized with 0.5% Triton

180

X-100 (TX-100). Following blocking, the cells were incubated with primary antibodies

181

(Abs) and subsequently with secondary Abs conjugated with Alexa Fluor 488, 568, or

182

647 (Molecular Probes). The following Abs were used as primary Abs: mouse anti-HA

183

mAb12-1G6 (24), mouse anti-HA Ab (Takara), rabbit anti-NA Ab (Sino Biological),

184

sheep anti-NA Ab (R&D Systems), rabbit anti-M1 polyclonal Ab (25), mouse anti-M2

185

Ab (Santa Cruz), mouse anti-NP mAb61A5 (26), mouse anti-CD59 Ab (Abcam), rabbit

186

anti-TGN46 Ab (Abcam), rabbit anti-ZO-3 Ab (Invitrogen), and rabbit anti-caveolin-1

187

Ab (Santa Cruz). For costaining with HA and M2 or vRNP, anti-HA mAb12-1G6 was

188

pre-labeled by using the Zenon Alexa Fluor 488 mouse IgG1 labeling kit (Invitrogen),

189

and the mouse anti-HA (Takara), anti-M2, and anti-NP Abs were similarly pre-labeled

190

with Alexa Fluor 568, as described previously (24). Nuclear staining was carried out

191

with TO-PRO-3 (Molecular Probes) or DAPI (Molecular Probes). The cells were

192

observed with a laser scanning confocal microscope (TCS-SP5II AOBS; Leica).

193

Confocal images were collected at 0.5-μm intervals along the Z-axis. Reconstitution of

194

an X-Z plane was processed using ImageJ software (27). Fifty antigen-positive cells

195

were observed in each experiment and patterns of antigen distribution were analyzed.

196

For quantitative analysis of HA and NA expression, polarized MDCK cells on

197

coverslips in 12-well plates (1×106 cells/well) were singly transfected with an HA

198

expression plasmid or cotransfected with the HA and an NA expression plasmids. After

199

fixation with 4% PFA, the cells were incubated with mouse anti-HA Ab (Takara) (for

200

HA on the apical PM). The cells were re-fixed with 4% PFA and were permeabilized

201

with 0.5% TX-100. The cells were incubated with rabbit anti-NA Ab (Sino Biological)

202

(for total NA) and subsequently with anti-mouse IgG conjugated with Alexa Fluor 647

203

and anti-rabbit IgG conjugated with Alexa Fluor 568. For total HA, the cells were

204

re-fixed with 4% PFA and were blocked with nonspecific mouse Ig. The cells were

205

finally incubated with anti-HA mAb12-1G6 pre-labeled by using the Zenon Alexa Fluor

206

488 mouse IgG labeling kit (Invitrogen). Nuclear staining was carried out with DAPI

207

(Molecular Probes). Confocal images were collected at 0.5-μm intervals along the

208

Z-axis. In each cell, the sum of intensity values of a Z-stack was calculated as a

209

Z-projection image by “sum slices” command of ImageJ. In each acquired channel,

210

single cell area and mean fluorescence intensities (MFI, arbitrary unit) were measured.

211

The MFI of Alexa Fluor 488 and 568 channels were evaluated as total expression levels

212

of HA and NA in each cell, respectively. The total fluorescence intensity (MFI × area)

213

of HA divided by that of NA was evaluated as the expression ratio of HA-to-NA. The

214

cells (39-45 cells) were subjected to analysis of HA distribution patterns (either apical

215

PM alone or PMs plus cytoplasmic compartments). Sparse MDCK cells (1×105

216

cells/well) were also used in Fig. 3C.

217 218

TX-100 solubilization analysis and coimmunoprecipitation

219

293T cells were seeded into 12-well plates (5×105 cells/well) or

220

10-cm-diameter dishes (5×106 cells/dish). The cells were singly transfected with HA

221

and NA expression plasmids or cotransfected with a combination of HA and NA, HA

222

and M1, or HA and GFP-CD59 expression plasmids, respectively. At 24 hpt, the cells

223

were resuspended with cold TNE buffer (50 mM Tris-HCl [pH 7.5], 1 mM EDTA, and

224

150 mM NaCl) containing 1 mM DTT and protease inhibitor complete mini cocktail

225

(Roche). After brief sonication, the samples were treated with 1% TX-100 at 4°C or

226

37°C for 30 min and were centrifuged at 17,400×g for 30 min at 4°C to separate the

227

soluble and insoluble fractions. For TX-100 solubilization analysis, both the

228

supernatants and pellets were adjusted to be the same volume with TNE buffer and

229

analyzed by Western blotting. For coimmunoprecipitation, the supernatants were mixed

230

with 10 μg of anti-HA mAb12-1G6 and were incubated at 4°C for 90 min. The

231

supernatants were subsequently mixed with pre-blocked Protein G-Sepharose-beads

232

(GE Healthcare) and were incubated at 4°C for 60 min. After washing with TNE

233

containing 0.1% TX-100 three times, immunoprecipitates were eluted from the beads by

234

boiling with SDS sample buffer and were analyzed by Western blotting with sheep

235

anti-NA Ab (R&D Systems), rabbit anti-M1 polyclonal Ab (25). GFP-CD59 was

236

detected with mouse anti-GFP Ab (Sigma).

237

238

In situ proximity ligation assay (PLA)

239

In situ PLA was performed according to the manufacturer’s instructions (Olink

240

Biosciences). Briefly, polarized MDCK cells were either transfected with combinations

241

of protein expression plasmids or were infected with virus. At 9 hpt or 3.5 or 5 h

242

post-infection (hpi), the cells were fixed with 4% PFA, and permeabilized with 0.5%

243

TX-100. After blocking, the cells were incubated with mouse anti-HA mAb12-1G6,

244

rabbit anti-NA Ab, mouse anti-GFP Ab or rabbit caveolin-1 Ab (Santa Cruz) for 1 h at

245

room temperature. After being washed, the cells were incubated with two

246

species-specific secondary antibodies conjugated with unique oligonucleotides (rabbit

247

PLUS and mouse MINUS) as PLA probes. Phospholyrated connector oligonucleotides

248

were hybridized to the PLA probes. If the PLA probes are present in close proximity

249

(

Influenza A virus hemagglutinin and neuraminidase mutually accelerate their apical targeting through clustering of lipid rafts.

In polarized epithelial cells, influenza A virus hemagglutinin (HA) and neuraminidase (NA) are intrinsically associated with lipid rafts and target th...
6MB Sizes 0 Downloads 2 Views