JVI Accepted Manuscript Posted Online 17 February 2016 J. Virol. doi:10.1128/JVI.02878-15 Copyright © 2016, American Society for Microbiology. All Rights Reserved.

1

Linear epitopes in A27 are targets of protective antibodies induced

2

by vaccination against smallpox

3 4

Running title: Protective linear epitopes of vaccinia virus A27

5 6

Thomas Kaever1, Michael H. Matho2, Xiangzhi Meng3, Lindsay Crickard1, Andrew Schlossman2,

7

Yan Xiang3, Shane Crotty1,4, Bjoern Peters1, Dirk M. Zajonc2,5^

8 9 10 11 12 13 14 15 16

1

Division of Vaccine Discovery, 2Division of Cell Biology, La Jolla Institute for Allergy and

Immunology (LJI), La Jolla, CA 92037, USA. 3

Department of Microbiology and Immunology, University of Texas Health Science Center at

San Antonio, San Antonio, TX 78229. 4

Department of Medicine, University of California, San Diego School of Medicine, La Jolla, CA

92037, USA 5

Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University,

9000 Ghent, Belgium

17 18

^ Corresponding author:

19

Dirk Zajonc

20 21

Abstract: 225 words

22

Full text: 7,496 words

1

23

Abstract

24

Vaccinia virus (VACV) A27 is a target for viral neutralization and part of the Dryvax

25

smallpox vaccine. A27 is one of the three glycosaminoglycan (GAG) adhesion molecules and

26

binds to heparan sulfate. To understand the function of anti-A27 antibodies, especially their

27

protective capacity and their interaction with A27, we generated and subsequently characterized

28

7 murine monoclonal antibodies (mAbs), which fell into 4 distinct epitope groups. Three groups

29

(I, III and IV) bound to linear peptides, while group II only bound to VACV lysate and

30

recombinant A27, suggesting it recognized a conformational and discontinuous epitope. Only

31

group I antibodies neutralized MV in a complement-dependent manner and protected against

32

VACV challenge, while a group II mAb protected partially but did not neutralize. The epitope

33

for group I mAbs was mapped to a region adjacent to the GAG binding site and suggest that

34

group I mAbs could potentially interfere with cellular adhesion of A27. We have further

35

determined the crystal structure of the neutralizing group I mAb 1G6, as well as the non-

36

neutralizing group IV mAb 8E3 bound to the corresponding linear epitope-containing peptides.

37

Both light and heavy chains of the antibodies are important in binding their antigens. For both

38

antibodies the L1 loop seems to dominate the overall polar interactions with the antigen, while

39

for mAb 8E3, the light chain generally appears to make more contacts with the antigen.

40 41

Importance

42

Vaccinia virus is a powerful model to study antibody responses upon vaccination, since

43

its use as the smallpox vaccine led to the eradication of one of the world’s greatest killers. The

44

immunodominant antigens that elicit the protective antibodies are known, yet for many of these 2

45

antigens little information about their precise interaction with antibodies is available. In an

46

attempt to better understand the interplay between the antibodies and their antigens, we have

47

studied a panel of anti A27 antibodies from generation, functional characterization to the

48

interaction with the epitope using X-ray crystallography. We identified one protective antibody

49

that binds adjacent to the heparan sulfate binding site of A27, likely affecting ligand binding.

50

The analysis of antibody-antigen interaction supports a model in which antibodies that can

51

interfere with the functional activity of the antigen are more likely to confer protection than those

52

that bind at the extremities of the antigen.

53 54 55

Introduction Inoculation with vaccinia virus (VACV) elicits neutralizing antibodies against major

56

antigens, including A27, A33, B5, D8, H3, and L1 on both the extracellular enveloped virus (EV)

57

and the intracellular mature virion (MV or IMV), conferring protection against smallpox (2, 6, 7,

58

16, 26). As a result, wide-spread vaccination against smallpox (variola virus, VARV) led to the

59

first eradication of a viral pathogen from nature (11). Among the major immunodominant

60

antigens of the IMV, A27, H3, and D8 are adhesion molecules that bind to the

61

glycosaminoglycans heparan sulfate (A27 and H3) and chondroitin sulfate (D8) (13, 14, 18, 20).

62

We have previously shown that anti-D8 antibodies can prevent binding of D8 to chondroitin

63

sulfate. Besides its binding to heparan sulfate, little is known about the function of H3 (18).

64

However, human antibodies that target H3 in combination with those that target B5 provide

65

significantly better protection than either antibody by itself and are promising for the treatment

66

of smallpox infections in human (22). Since the general human population lacks protection

3

67

against smallpox due to the cessation of smallpox vaccination, protective antibodies can be used

68

to treat infected patients. While neutralizing anti-A27 antibodies protect against infection, they

69

only represent a minor component of the Dryvax vaccine induced immune response (10).

70

A27 is a homo-trimeric extracellular protein that is attached to the viral membrane by

71

binding to the transmembrane protein A17 through its C-terminal leucine zipper domain

72

(residues 80-101). The GAG binding site is located at the N-terminus, downstream of the signal

73

sequence (residues 21-30) (12, 30). The central region of A27 consists of a coiled coil domain

74

(residues 43-84), which is used to interact with the membrane fusion suppressor protein A26

75

through intermolecular disulfide bond formation (Cys71, Cys72). The crystal structure of an N-

76

terminal fragment of A27, containing the heparan sulfate binding site and coiled coil domain

77

(residues 21-84) had recently been determined, however only the central fragment (residues 47-

78

84) is ordered, suggesting flexibility of the N-terminal GAG binding domain (5). The A27

79

structure illustrates the complexity and antiparallel nature of the A27 homo-trimer, yet structural

80

information about the N-terminal and C-terminal extremities are missing.

81

In this study we have produced a panel of anti-A27 antibodies through immunizing mice

82

with VACV. We have identified 4 antibody groups based on cross-blocking experiments and

83

identified the epitope using a peptide/protein ELISA. Group I, II, and IV antibodies recognized

84

both VACV lysate as well as synthetic peptides, suggesting that the epitope of these antibodies

85

can be recapitulated using linear peptides. We have further determined the crystal structure of a

86

protective antibody 1G6 (group I), and a non-protective antibody 8E3 (group IV) in complex

87

with their respective linear peptide epitopes, which are located at the N- and C-terminal

88

extremities of A27, shedding light onto the structural basis of A27 recognition.

4

89

Materials and Methods

90

Viruses and antibodies. VACVWR stocks were grown on HeLa cells in T175 flasks and

91

infecting at a multiplicity of infection of 0.5. Cells were harvested at 60 h and virus was isolated

92

by rapidly freeze-thawing the cell pellet three times in a volume of 2.3 ml RPMI plus 1% fetal

93

calf serum (FCS). Subsequently, cell debris was removed by centrifugation. Clarified supernatant

94

was frozen at -80°C as stock. VACVWR stocks were titered on Vero cells (~2 x 108 PFU/ml).

95

VACVACAM2000 was obtained from the CDC. Monoclonal antibodies used in the study are: anti-

96

H3 #41, anti-D8 Ab12.1, anti-L1 M12B9, anti-A27 1G6, 12G2, 8H10, 6F11, 4G5, 12C3 and 8E3.

97

Hybridoma generation and characterization.

98

The hybridomas were generated as described previously (32). In brief, a six-week old BALB/c

99

mouse was infected intranasally with 5 × 103 plaque-forming-unit (PFU) of VACV WR. Seven

100

weeks after the infection, the mouse was injected intravenously with 7 × 107 PFU of UV-

101

inactivated WR virus. Three days afterwards, the spleen of the mouse was harvested for

102

hybridoma generation. Hybridomas that secret anti-A27 antibodies were identified with

103

immunofluorescence assays of HeLa cells infected with wild-type or A27-deletion VACV strains

104

(31).

105

chromatography and purity was assessed by reducing and non-reducing SDS PAGE. All

106

experiments were performed using purified antibodies.

107

A27 expression and purification.

108

A27L with a C-terminal hexahistidine tag was cloned into pET15b (Invitrogen) and transformed

109

into CodonPlus BL21 cells (Agilent). After culture growth reached an OD600 of 0.6, A27

110

expression was induced by induction with 1mM IPTG at 37 °C for 4 h. Cells from typically 1 L

IgG’s were purified from hybridoma culture media using protein G affinity

5

111

were spun down (10 min x 4000 g) and resuspended in 50 ml lysis buffer (50 mM Tris pH 8.0, 5

112

mM EDTA). Cells were sheared with a microfluidizer (3 rounds at 1400 bars, Microfluidics) and

113

crude lysate was clarified by centrifugation (1 h x 50,000 g). Soluble A27 was purified by ion

114

metal affinity chromatography (IMAC) using a HisTrap 5 ml column (GE Healthcare).

115

Supernatant was passed through the HisTrap column and washed with three column volumes (15

116

ml) of wash buffer (50 mM Tris pH 8.0, 300 mM NaCl, 20 mM imidazole). After a final wash at

117

50 mM imidazole, A27 was eluted with the same buffer containing ~ 250 mM imidazole and

118

subsequently dialyzed twice against 10 mM tris pH8.0, 200 mM NaCl prior to size-exclusion

119

chromatography (SEC) for characterization.

120

Protein ELISA. Flat-bottom 96-well microtiter plates were coated with 100 µl of recombinant

121

VACV A27 protein (either C-terminally truncated A27(16-100) or full length A27(16-110) at

122

1 mg/ml, diluted in PBS overnight at 4°C (ThermoScientific Pierce) and washed with washing

123

buffer (PBS, pH 7.2, plus 0.05% Tween 20). Subsequently, plates were blocked with blocking

124

buffer (PBS, pH 7.2, plus 1% BSA plus 0.1% Tween 20) for 2 h at room temperature (RT).

125

Plates were washed and incubated with purified mAb at 10 µg/ml for 90 min at RT. Plates were

126

washed, and the bound mAb was detected by adding a streptavidin-HRP-conjugated secondary

127

antibody to mouse immunoglobulin G (Invitrogen) and incubated for 60 min at RT, followed by

128

OPD substrate (Sigma-Aldrich).

129

Cross-Blocking ELISA. Recombinant A27(16-110) was prepared at 0.5 µg/ml and used to coat

130

Nunc Polysorbent flat-bottom 96-well plates with 100 µl per well. Plates were incubated

131

overnight at 4°C and subsequently washed four times with PBS plus 0.05% Tween 20. 100 µl of

132

blocking buffer (PBS + 10% fetal bovine serum) was added to each well of the plate and the

133

plate was blocked for 90 min at room temperature. Blocking buffer was discarded, and 100 µl of 6

134

purified and unmodified antibodies of interest (at 10 µg/ml) were added to the plate and

135

incubated for 90 min to allow for binding to recombinant A27. Horseradish peroxidase (HRP)-

136

conjugated antibodies of interest (Innova Biosciences Lightning-Link HRP conjugation kit) were

137

prepared at 0.5 µg/ml and added to the plates for 20 min without prior washing of the plate. The

138

ability of the conjugated antibody to bind to A27 in the presence of a pre-bound antibody was

139

assessed using an optical assay. The plates were developed using o-phenylenediamine (OPD),

140

and optical density (OD) at 490 nm was read on a SpectraMax 250 (Molecular Devices).

141

Flow Cytometry-based in vitro neutralization. Vero E6 cells (1x105 cells/well) were seeded in

142

96-well Costar plates (Corning Inc., Corning, NY) and incubated for 5 h to adhere. Subsequently,

143

cells were infected with 12.5 µL purified VACV-GFP at 1x106 PFU/mL (final PFU of 1.25x104)

144

and 12.5 µL mAbs at 80 µg/mL (final concentration of 20 µg/mL) for 12 h at 37°C and 5% CO2

145

in a total volume of 50 µL in the presence (2% final concentration) or absence of sterile baby

146

rabbit complement (CEDARLANE). Samples were prepared in duplicates. Cells were

147

subsequently tested using flow cytometry as described previously (1, 15).

148

Vaccinia intravenous infection protection studies. To infect mice, 1x105 PFU of

149

VACVACAM2000 were injected retro-orbitally. After infecting animals on day 0, weights were

150

taken on day 0 for the initial body weight measurement. Beginning three days post-infection,

151

body weights were taken every other day. Body weights were recorded until the animal reached

152

75% of initial body weight, or if other external health variables became present for which

153

euthanasia was the only humane course of action. Clinical score, a composite score of the pox

154

lesion abundance on the four paws plus the tail, was evaluated as described (22). For A27 mAb

155

protection studies, mice were inoculated i.p. with 100 µg of antibodies 1 day before infection.

156

Control mice received PBS. An additional group received anti-H3 #41 as positive control. 7

157

Animal husbandry and experimental procedures were approved by the Department of Laboratory

158

Animal Care and the Animal Care Committee of the La Jolla Institute

159

Epitope Mapping by Peptide ELISA. Overlapping 20-mer peptides for the A27 antigen were

160

synthesized (AnaSpec) and tested for mAb binding using an ELISA. Flat-bottom 96-well

161

microtiter plates were coated with 100 µl of NeutrAvidin biotin-binding protein (1 mg/ml),

162

diluted in PBS overnight at 4°C (ThermoScientific Pierce) and washed with washing buffer (PBS,

163

pH 7.2, plus 0.05% Tween 20). Subsequently, plates were blocked with blocking buffer (PBS,

164

pH 7.2, plus 1% BSA plus 0.1% Tween 20) for 2 h at room temperature (RT). Plates were

165

incubated with 100 µl of overlapping linear biotinylated peptides (200 ng/ml) in blocking buffer

166

for 90 min at RT. Plates were washed and incubated with purified mAb at 10 µg/ml for 90 min at

167

RT. Plates were washed, and the bound mAb was detected by adding a streptavidin-HRP-

168

conjugated secondary antibody to mouse immunoglobulin G (Invitrogen) and incubated for 60

169

min at RT, followed by OPD substrate (Sigma-Aldrich).

170

Peptide Truncation and Alanine Scan. Variant peptides with N- or C-terminal truncations

171

and/or alanine substitutions were tested for their ability to block binding to the parent 20-mer

172

peptides in ELISA. In case the peptide in question contained an alanine, it was substituted for a

173

serine instead. 96-well plates were coated with 100 µL NeutrAvidin per well at a concentration

174

of 0.5 µg/mL. Plates were incubated overnight at 4°C and washed 4 times with PBS plus 0.05%

175

Tween-20. 100 µL of blocking buffer (PBS + 10% FBS) were added to the plates and incubated

176

for 90 min at 4°C. Subsequently, blocking buffer was discarded, 100 µL of biotinylated 20-mer

177

peptides were added to the plate at 200 ng/mL and incubated for 90 min at 4°C. Simultaneously,

178

selected antibodies were incubated with variant peptides. We used 30 µL/well of mAb at 600

179

ng/mL and incubated it with 30 µL/well of alanine-modified peptides at 100 µg/mL for 90 min at 8

180

4°C. After washing the plates, 50 µL of the antibody/alanine peptide mix was added to plate

181

bound peptides and incubated for 20 min at 4°C. Plates were washed and 100 µL/well of

182

secondary goat anti-mouse anti IgGγ HRP (1:1000 diluted in blocking buffer) were added and

183

incubated for 90 min at 4°C. A final wash step was performed and plates were developed using

184

o-phenylenediamine and OD at 490nm was read on a SpectraMax 250 (Molecular Device).

185

Antibody sequencing. Total RNA from 300 µL hybridoma cells in solution was isolated using

186

the NucleoSpin® RNA II kit according to manufacturer’s instructions (MACHEREY-NAGEL).

187

cDNA was amplified using the OneStep RT-PCR kit (Qiagen). The reverse transcription PCR

188

was performed using primers 5’MsVHE and 3’Cy1 (for isotype IgG1 mAb 6F11), 3’Cy2c outer

189

(for isotype IgG2a mAbs 1G6, 12G2, 12C3 and 4G5) or 3’Cy2b outer (for isotype IgG2b mAb

190

8H10) for the heavy chains, primers 5’mVkappa and 3’mCĸ for the kappa light chains (1G6,

191

12G2, 8H10, 6F11, 12C3 and 4G5) and primers 5’mVλ1/2 and 3’mCλ outer for the lambda light

192

chain (8E3) (29). The cycling profile was slightly modified from manufacturer’s

193

recommendations and set up as follows: 1 cycle of 30 min at 50°C and 15 min at 95°C; 40 cycles

194

of 30 s at 94°C, 45 s at 60°C (for heavy chains) / 58°C (for light chains), and 55 s at 72°C;

195

followed by 1 cycle of 10 min at 72°C and a 12°C cool down. PCR products were verified by gel

196

electrophoresis with a ~500 bp product for heavy chains and ~450 bp product for light chains.

197

Afterwards, PCR products were purified using the QIAquick PCR Purification Kit (Qiagen) and

198

then sequenced by Invitrogen (provided with the respective 5’ primer for heavy and light chains).

199

Sequences include V-D-J regions for heavy chains and V-J regions for light chains. Finally,

200

antibody germ lines were determined using IMGT’s V-Quest service (3).

201

Peptide-Fab complex preparation. Purified mAbs 1G6 and 8E3 (1mg/mL in 50 mM NaOAc

202

pH5.5) were incubated with 4% (1G6) and 2% (8E3) (w/w) activated papain (Sigma #P3125) 9

203

and for 4h at 37ºC in 1X digestion buffer. Papain was activated by incubating 20.80 µl papain

204

with 100 µl 10X digestion buffer (1M NaOAc pH 5.5, 12 mM EDTA) and 100µl cysteine (12.2

205

mg/ml) in a total volume of 1ml for 15 min at 37ºC. The papain digestion was stopped by adding

206

20 mM Iodoacetamide (IAA). Digestion mixtures were dialyzed against PBS for subsequent

207

protein A purification to remove undigested IgG and Fc. The protein A flow-through containing

208

Fab’s were concentrated and purified by size exclusion chromatography on a Superdex S200

209

GL10/300 (GE Healthcare), using 50 mM Hepes, pH7.5, 150mM NaCl as running buffer. Fab’s

210

were incubated with 2x molar excess of corresponding A27 peptides for 1 hour at 4˚C and

211

concentrated using 30 kDa centrifugal filtration units to remove unbound peptides.

212

Crystallization and structure determination. Crystals of the 1G6/A2731-40 complex were

213

grown over several days at 22°C by sitting drop vapor diffusion while mixing 0.5 µl protein (5.2

214

mg/ml) with 0.5 µl precipitant (20 % PEG 4000, 200 sodium phosphate dibasic). Crystals were

215

flash-cooled at 100 K in mother liquor containing 20% glycerol. Crystals of the 8E3/A27101-110

216

complex were grown over several days at 4°C by sitting drop vapor diffusion while mixing 0.5

217

µl protein (14 mg/ml) with 0.5 µl precipitant (10 % PEG 3000, 200 mM magnesium chloride,

218

100 mM cacodylate pH 6.5). Crystals were flash-cooled at 100 K in mother liquor containing 20%

219

glycerol. Diffraction data were collected at the Stanford Synchrotron Radiation Laboratory

220

(SSRL) beamline 11-1 (1G6 Fab) and Advanced Light source beamline 5.0.1 (8E3 Fab),

221

processed with iMosflm and SCALA as part of ccp4 (4, 17). Crystal structure of 8E3 was

222

determined by molecular replacement using PHASER (23) and the Fab of a quorum-quenching

223

antibody (PDB code 2NTF), separated in constant and variable domain. The structure of 1G6

224

was obtained similarly by MR, using the PDB coordinated of 8E3 with the CDR loops removed.

225

The model was rebuilt into σA-weighted 2Fo–Fc and Fo–Fc difference electron density maps 10

226

using the program COOT (8). Peptides were manually built in COOT during later stages of

227

refinement. The 1G6/A2731-40 structure was refined to 1.95 Å to an Rcryst and Rfree of 19.8% and

228

23.6%, respectively. The 8E3/A27101-110 structure was refined to 2.27 Å to an Rcryst and Rfree of

229

20.3% and 22.4%, respectively The quality of the models were examined with the program

230

Molprobity (19).

231 232

Results

233

Generation of anti-A27 mAbs

234

Hybridomas were generated from a mouse that had been infected with a sub-lethal dose

235

of VACV. Among them, 12 secreted antibodies reacted with the wild-type VACV but not with

236

an A27-deletion VACV mutant in an immunofluorescence assay of infected HeLa cells. We

237

decided to further characterize seven of those antibodies (1G6 [IgG2a], 12G2 [IgG2a], 8H10

238

[IgG2b], 6F11 [IgG1], 12C3 [IgG2a], 4G5 [IgG2a] and 8E3 [IgG2a]). Out of these, 6 antibodies

239

bound C-terminally truncated A27(16-100) protein in ELISA, whereas the other antibody (8E3)

240

did not bind A27 (Figure 1A). To assess whether 8E3 binds to the C-terminal extremity of A27,

241

we prepared full-length A27(16-110) and repeated the ELISA with four selected mAbs,

242

including 8E3. All tested antibodies were found to bind the new construct, suggesting that 8E3

243

binds to the C-terminal extremity of A27 (Figure 1B).

244

Cross-blocking ELISA was performed, and antibodies 1G6, 12G2 and 8H10 clustered

245

into one distinct cluster (group I). Additionally, 4G5 and 12C3 formed another, distinct cluster

246

(group III). 6F11 and 8E3 could not be cross-blocked by any of the other mAbs and were

247

assigned group II and IV, respectively (Figure 2).

11

248 249

Group I anti-A27 mAbs neutralize MV in a complement-dependent manner

250

We then tested the ability of the anti-A27 mAbs to interfere with VACV MV infection in

251

an in vitro neutralization assay (Figure 3). Vero E6 cells were incubated overnight with purified

252

VACVWR MV expressing a green fluorescent protein (GFP), in the presence or absence of

253

antibody and complement. The samples were evaluated the following day using a flow

254

cytometer. Group I mAbs (1G6, 12G2 and 8H10) neutralized more than 90% of the viruses at 20

255

µg/ml in the presence of complement. In contrast, group II, III and IV mAbs only neutralize the

256

virus by 20% or less in the presence of complement (Figure 3, bottom). Except for mAb 6F11

257

(IgG1), all antibodies were able to bind complement (IgG2a and IgG2b). None of the tested

258

mAbs was able to neutralize in the absence of complement (Figure 3, top). Anti-L1 mAb

259

M12B9 (15), an antibody know to strongly neutralize (>90%) in a complement-independent

260

manner, was used as positive control.

261 262

Group I anti-A27 mAb protects against vaccinia virus infection

263

Next, anti-A27 mAbs were tested in an in vivo vaccinia protection system. SCID mice

264

were infected with 105 PFU ACAM2000 retro-orbitally (Figure 4). In this model, 1G6 (group I),

265

6F11 (group II) and 8E3 (group IV) were tested for their ability to protect against moribundity,

266

weight loss and pox lesions (‘Clinical Score’) (24). Animals were euthanized at 75% initial body

267

weight (end point criteria). Anti-H3 #41 was used as positive control; at day 49 it provided good

268

protection against weight loss (p=0.0024) (Figure 4A) and mortality (p=0.0008) (Figure 4B)

269

compared to control mice receiving no mAb. Anti-H3 #41 also provided robust protection

12

270

against pox lesions (p90%) w/ complement strong (>90%) w/ complement weak (

Linear Epitopes in Vaccinia Virus A27 Are Targets of Protective Antibodies Induced by Vaccination against Smallpox.

Vaccinia virus (VACV) A27 is a target for viral neutralization and part of the Dryvax smallpox vaccine. A27 is one of the three glycosaminoglycan (GAG...
5MB Sizes 1 Downloads 8 Views